Network


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

Hotspot


Dive into the research topics where Jianbo Fan is active.

Publication


Featured researches published by Jianbo Fan.


Biochemical and Biophysical Research Communications | 2016

α-Melanocyte stimulating hormone attenuates dexamethasone-induced osteoblast damages through activating melanocortin receptor 4-SphK1 signaling

Shiguang Guo; Yue Xie; Jianbo Fan; Feng Ji; Shouguo Wang; Haodong Fei

Long-term glucocorticoid (GC) usage may cause non-traumatic femoral head osteonecrosis. Dexamethasone (Dex) is shown to exert potent cytotoxic effect to osteoblasts. Here, we investigated the potential activity of α-melanocyte stimulating hormone (α-MSH) against the process. Our data revealed that pretreatment of α-MSH significantly inhibited Dex-induced apoptosis and necrosis in both osteoblastic-like MC3T3-E1 cells and primary murine osteoblasts. Melanocortin receptor 4 (MC4R) acts as the receptor of α-MSH in mediating its actions in osteoblasts. The MC4R antagonist SHU9119, or shRNA-mediated knockdown of MC4R, almost abolished α-MSH-induced activation of downstream signalings (Akt and Erk1/2) and its pro-survival effect in osteoblasts. Further studies showed that α-MSH activated MC4R downstream sphingosine kinase 1 (SphK1) and increased cellular sphingosine-1-phosphate (S1P) content in MC3T3-E1 cells and primary murine osteoblasts, which were blocked by SHU9119 or MC4R shRNAs. SphK1 inhibition by the its inhibitor N,N-dimethylsphingosine (DMS), or SphK1 knockdown by targeted-shRNAs, largely attenuated α-MSH-mediated osteoblast protection against Dex. Together, these results suggest that α-MSH alleviates Dex-induced damages to cultured osteoblasts through activating MC4R-SphK1 signaling.


Journal of the Neurological Sciences | 2011

Overexpression of glucose-regulated protein 94 after spinal cord injury in rats.

Dawei Xu; Shengyu Cui; Yuyu Sun; Guofeng Bao; Weidong Li; Wei Liu; Xinhui Zhu; Jianbo Fan; Youhua Wang; Zhiming Cui

Glucose-regulated protein (GRP) 94 is a member of the stress protein family, which is localized in the endoplasmic reticulum (ER). Spinal cord injury (SCI) induced ER stress that results in apoptosis. However, the role of GRP94 in injury of the central nervous system remains unknown. In this study, we performed SCI in adult rats and investigated acutely the protein expression and cellular localization of GRP94 in the spinal cord. Western blot analysis revealed that GRP94 was low in normal spinal cord. It rose at 6h after SCI, peaked at 1 day, remained for another 3 days, then declined to basal levels at 5 days after injury. Immunohistochemistry further confirmed that GRP94 immunoactivity was expressed at low levels in gray matter and white matter in normal condition and increased after SCI. Double immunofluorescence staining showed that GRP94 was co-expressed with NeuN (neuronal marker), and GFAP (astroglial marker). In addition, caspase-12, caspase-3 and phospho-c-Jun NH2-kinase (p-JNK) levels increased at 6h, peaked at 1day, and then gradually reduced to normal levels for 2 weeks after SCI by western blot analysis. Co-localization of GRP94/caspase-12 and GRP94/p-JNK was detected in neurons and glial cells. Taken together, these data suggest GRP94 involvement in the injury response of the adult spinal cord of the rats.


Molecular and Cellular Biochemistry | 2015

EGFR-AKT-mTOR activation mediates epiregulin-induced pleiotropic functions in cultured osteoblasts

Jianbo Fan; Wei Liu; Xinhui Zhu; Kun Yuan; Dawei Xu; Jiajia Chen; Zhiming Cui

Epidermal growth factor (EGF) receptor (EGFR) emerges as an essential molecule for the regulating of osteoblast cellular functions. In the current study, we explored the effect of epiregulin, a new EGFR ligand, on osteoblast functions in vitro, and studied the underlying mechanisms. We found that epiregulin-induced EGFR activation in both primary osteoblasts and osteoblast-like MC3T3-E1 cells. Meanwhile, epiregulin activated AKT-mammalian target of rapamycin (mTOR) and Erk-mitogen-activated protein kinase (MAPK) signalings in cultured osteoblasts, which were blocked by EGFR inhibitor AG1478 or monoclonal antibody against EGFR (anti-EGFR). Further, in primary and MC3T3-E1 osteoblasts, epiregulin promoted cell proliferation and increased alkaline phosphatase activity, while inhibiting dexamethasone (Dex)-induced cell death. Such effects by epiregulin were largely inhibited by AG1478 or anti-EGFR. Notably, AKT-mTOR inhibitors, but not Erk inhibitors, alleviated epiregulin-induced above pleiotropic functions in osteoblasts. Meanwhile, siRNA depletion of Sin1, a key component of mTOR complex 2 (mTORC2), also suppressed epiregulin-exerted effects in MC3T3-E1 cells. Together, these results suggest that epiregulin-induced pleiotropic functions in cultured osteoblasts are mediated through EGFR-AKT-mTOR signalings.


Journal of Molecular Neuroscience | 2013

MCM7 Expression Is Altered in Rat After Spinal Cord Injury

Jiajia Chen; Zhiming Cui; Weidong Li; Aiguo Shen; Guanhua Xu; Guofeng Bao; Yuyu Sun; Lingling Wang; Jianbo Fan; Jinlong Zhang; Longfei Yang

Minichromosome maintenance protein 7 (MCM7), a member of the minichromosome maintenance protein family, is essential for eukaryotic DNA replication initiation and the early stage of the elongation process. MCM7 participates in the cell cycle control of genome duplication. While it is ubiquitously expressed in all tissues, the biological function of MCM7 in the central nervous system is still with limited acquaintance. In the present study, we performed a spinal cord injury (SCI) model in adult rats. Western blotting indicated a marked alteration of MCM7 after SCI. Immunohistochemistry analysis revealed a wide distribution of MCM7 in the spinal cord. Double immunofluorescence staining showed that MCM7 immunoreactivity was increased predominantly in neurons, astrocytes, and microglia after SCI. We also examined the expression profiles of active caspase-3, proliferating cell nuclear antigen (PCNA), and Ki67, whose changes were correlated with the expression profiles of MCM7. Moreover, colocalization of MCM7/active caspase-3 was detected in neuronal nuclei (NeuN), and colocalization of MCM7/PCNA was detected in NeuN, glial fibrillary acidic protein, and CD11b, respectively. Our results suggest that MCM7 might be implicated in the apoptosis of neuron and proliferation of astrocytes and microglia after SCI.


Oncotarget | 2017

microRNA-25 targets PKCζ and protects osteoblastic cells from dexamethasone via activating AMPK signaling

Jianbo Fan; Wei Liu; Xinhui Zhu; Hong Yi; Shengyu Cui; Jian-ning Zhao; Zhiming Cui

AMP-activated protein kinase (AMPK) activation could protect osteoblasts from dexamethasone (Dex). This study aims to provoke AMPK activation via microRNA downregulation of its negative regulator protein kinase C ζ (PKCζ). Results show that microRNA-25-5p (miR-25-5p) targets PKCζs 3’ untranslated regions (UTRs). Forced-expression of miR-25 downregulated PKCζ and activated AMPK in human osteoblastic cells (OB-6 and hFOB1.19 lines), which thereafter protected cells from Dex. Reversely, expression of antagomiR-25, the miR-25 inhibitor, upregulated PKCζ and inhibited AMPK activation, exacerbating Dex damages. Notably, PKCζ shRNA knockdown similarly activated AMPK and protected osteoblastic cells from Dex. AMPK activation was required for miR-25-induced osteoblastic cell protection. AMPKα shRNA or dominant negative mutation almost completely blocked miR-25-induced cytoprotection against Dex. Further studies showed that miR-25 expression increased NADPH activity and suppressed Dex-induced oxidative stress in osteoblastic cells. Such effects by miR-25 were abolished with AMPKα knockdown or mutation. Significantly, miR-25-5p level was increased in patients’ necrotic femoral head tissues, which was correlated with PKCζ downregulation and AMPK hyper-activation. These results suggest that miR-25-5p targets PKCζ and protects osteoblastic cells from Dex possibly via activating AMPK signaling.


Experimental Biology and Medicine | 2016

Epigallocatechin-3-gallate protects against tumor necrosis factor alpha induced inhibition of osteogenesis of mesenchymal stem cells.

Wei Liu; Jianbo Fan; Dawei Xu; Jie Zhang; Zhiming Cui

Anabolic bone accruement through osteogenic differentiation is important for the maintenance of physiological bone mass and often disrupted in various inflammatory diseases. Epigallocatechin-3-gallate, as an antioxidant and anti-inflammatory agent, has been suggested for potential therapeutic use in this context, possibly by the inhibition of bone resorption as well as the enhancement of bone formation through directly activating osteoblast differentiation. However, the reported effects of epigallocatechin-3-gallate modulating osteoblast differentiation are mixed, and the underlying molecular mechanism is still elusive. Moreover, there is limited information regarding the effects of epigallocatechin-3-gallate on osteogenic potential of mesenchymal stem cell in inflammation. Here, we examined the in vitro osteogenic differentiation of human mesenchymal stem cells. We found that the cell viability and osteoblast differentiation of human bone marrow-derived mesenchymal stem cells are significantly inhibited by inflammatory cytokine TNFα treatment. Epigallocatechin-3-gallate is able to enhance the cell viability and osteoblast differentiation of mesenchymal stem cells and is capable of reversing the TNFα-induced inhibition. Notably, only low doses of epigallocatechin-3-gallate have such benefits, which potentially act through the inhibition of NF-κB signaling that is stimulated by TNFα. These data altogether clarify the controversy on epigallocatechin-3-gallate promoting osteoblast differentiation and further provide molecular basis for the putative clinical use of epigallocatechin-3-gallate in stem cell-based bone regeneration for inflammatory bone loss diseases, such as rheumatoid arthritis and prosthetic osteolysis.


Inflammation | 2012

Elevated Expression of β1,4-Galactosyltransferase-I in Cartilage and Synovial Tissue of Patients with Osteoarthritis

Wei Liu; Zhiming Cui; Youhua Wang; Xinhui Zhu; Jianbo Fan; Guofeng Bao; Junjun Qiu; Dawei Xu

Osteoarthritis (OA) is considered a complex illness, characterized by cartilage degeneration, secondary synovial membrane inflammation, and subchondral bone sclerosis. Previous studies have shown β1,4-galactosylransferase-I (β1,4-GalT-I) to be a key inflammatory mediator that participates in the initiation and maintenance of inflammatory reaction in diseases. In the present study, we investigated the expression and possible biological function of β1,4-GalT-I in the cartilage and synovial tissue of OA patients. Cartilage and synovial tissue samples from OA patients and healthy controls were stained for β1,4-GalT-I. Reverse transcription-polymerase chain reaction was used to observe the expression of β1,4-GalT-I, and western blot was carried out for E-selectin. The interaction between β1,4-GalT-I and E-selectin was analyzed by double labeling immunohistochemistry and immunoprecipitation. The expression of β1,4-GalT-I increased in the cartilage and synovial tissue of OA patients compared with healthy controls. E-selectin was overexpressed and was correlated with β1,4-GalT-I in OA cartilage and synovial tissue. These data suggest that β1,4-GalT-I may play an important role in the inflammatory processes in cartilage and synovial tissue of patients with OA.


Oncotarget | 2017

SphK2 over-expression promotes osteosarcoma cell growth

Dawei Xu; Hao Zhu; Chengniu Wang; Wei Zhao; Genxiang Liu; Guofeng Bao; Daoran Cui; Jianbo Fan; Fei Wang; Huricha Jin; Zhiming Cui

It is needed to explore novel biological markers for early diagnosis and treatment of human osteosarcoma. Sphingosine kinase 2 (SphK2) expression and potential functions in osteosarcoma were studied. We demonstrate that SphK2 is over-expressed in multiple human osteosarcoma tissues and established human osteosarcoma cell lines. Silence of SphK2 by targeted-shRNAs inhibited osteosarcoma cell growth, and induced cell apoptosis. On the other hand, exogenous over-expression of SphK2 could further promote osteosarcoma cell growth. Notably, microRNA-19a-3p (“miR-19a-3p”) targets the 3′ UTR (untranslated region) of SphK2 mRNA. Remarkably, forced-expression of miR-19a-3p silenced SphK2 and inhibited osteosarcoma cell growth. In vivo, SphK2 silence, by targeted-shRNA or miR-19a-3p, inhibited U2OS tumor growth in nude mice. These results suggest that SphK2 could be a novel and key oncotarget protein for OS cell progression.


PLOS ONE | 2016

OSU53 Rescues Human OB-6 Osteoblastic Cells from Dexamethasone through Activating AMPK Signaling.

Dawei Xu; Wei Zhao; Xinhui Zhu; Jianbo Fan; Shengyu Cui; Yuyu Sun; Xiang Chen; Wei Liu; Zhiming Cui

Excessive dexamethasone (Dex) application causes osteoblast cell death, which could lead to osteoporosis or osteonecrosis. AMP-activated protein kinase (AMPK) activation is shown to protect osteoblasts/osteoblastic cells from Dex. In this report, we tested the potential effect of OSU53, a novel AMPK activator, in Dex-treated osteoblastic cells. We show that OSU53 activated AMPK signaling in human OB-6 osteoblastic cells. Further, Dex-induced osteoblastic OB-6 cell death and apoptosis were largely attenuated with pre-treatment with OSU53. OSU53 was more efficient than other known AMPK activators (A-769662 and Compound 13) in protecting OB-6 cells against Dex. AMPK activation is required for OSU53-induced actions in OB-6 cells. AMPKα shRNA knockdown or dominant-negative mutation (dn-AMPKα T172A) almost completely blocked OSU53-induced AMPK activation and OB-6 cell protection against Dex. Further studies showed that OSU53 increased NADPH (nicotinamide adenine dinucleotide phosphate) activity and alleviated Dex-induced oxidative stress in OB-6 cells. Such effects by OSU53 were again almost abolished with AMPKα shRNA or dn-AMPKα in OB-6 cells. Together, these results demonstrate that OSU53 protects osteoblastic cells from Dex possibly via activating AMPK-dependent signaling.


Neuropeptides | 2018

Up-regulation of TNF Receptor-associated Factor 7 after spinal cord injury in rats may have implication for neuronal apoptosis

Dawei Xu; Wei Zhao; Chengniu Wang; Hao Zhu; Mingqing He; Xinhui Zhu; Wei Liu; Fei Wang; Jianbo Fan; Chu Chen; Daoran Cui; Zhiming Cui

TNF receptor-associated factor 7 (TRAF7), is an E3 ubiquitin ligase for several proteins involved in the activation of TLR-dependent NF-kappaB signaling. TRAF7 links TNF receptor family proteins to signaling pathways, thus participates in regulating cell death and survival mediated by TNF family ligands. To date, the biological function of TRAF7 after spinal cord injury (SCI) is still with limited acquaintance. In this study, we have performed an acute SCI model in adult rats and investigated the dynamic changes of TRAF7 expression in the spinal cord. Our results showed that TRAF7 was up-regulated significantly after SCI, which was paralleled with the levels of the apoptotic protein active caspase-3. Immunofluorescent labeling showed that TRAF7 was co-localizated with active caspase-3 in neurons. To further investigate the function of TRAF7, an apoptosis model was established in primary neuronal cells. When TRAF7 was knocked down by specific short interfering RNA (siRNA), the protein levels of active caspase-3 and the number of apoptotic primary neurons were significantly decreased in our study. Taken together, our findings suggest that the change of TRAF7 protein expression plays a key role in neuronal apoptosis after SCI.

Collaboration


Dive into the Jianbo Fan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge