Network


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

Hotspot


Dive into the research topics where Fanxia Shen is active.

Publication


Featured researches published by Fanxia Shen.


Cell Stem Cell | 2010

MicroRNA-9 Coordinates Proliferation and Migration of Human Embryonic Stem Cell-Derived Neural Progenitors

Celine Delaloy; Lei Liu; Jin-A Lee; Hua Su; Fanxia Shen; Guo-Yuan Yang; William L. Young; Kathy N. Ivey; Fen-Biao Gao

Human pluripotent stem cells offer promise for use in cell-based therapies for brain injury and diseases. However, their cellular behavior is poorly understood. Here we show that the expression of the brain-specific microRNA-9 (miR-9) is turned on in human neural progenitor cells (hNPCs) derived from human embryonic stem cells. Loss of miR-9 suppressed proliferation but promoted migration of hNPCs cultured in vitro. hNPCs without miR-9 activity also showed enhanced migration when transplanted into mouse embryonic brains or adult brains of a mouse model of stroke. These effects were not due to precocious differentiation of hNPCs. One of the key targets directly regulated by miR-9 encodes stathmin, which increases microtubule instability and whose expression in hNPCs correlates inversely with that of miR-9. Partial inhibition of stathmin activity suppressed the effects of miR-9 loss on proliferation and migration of human or embryonic rat neural progenitors. These results identify miR-9 as a novel regulator that coordinates the proliferation and migration of hNPCs.


Annals of Neurology | 2010

Endothelial progenitor cell transplantation improves long‐term stroke outcome in mice

Yongfeng Fan; Fanxia Shen; Tim Frenzel; Wei Zhu; Jianqin Ye; Jianrong Liu; Yongmei Chen; Hua Su; William L. Young; Guo-Yuan Yang

Endothelial progenitor cells (EPCs) play an important role in tissue repairing and regeneration in ischemic organs, including the brain. However, the cause of EPC migration and the function of EPCs after ischemia are unclear. In this study, we demonstrated the effects of EPCs on ischemic brain injury in a mouse model of transient middle cerebral artery occlusion (tMCAO).


Journal of Cerebral Blood Flow and Metabolism | 2008

Interleukin-6 stimulates circulating blood-derived endothelial progenitor cell angiogenesis in vitro

Yongfeng Fan; Jianqin Ye; Fanxia Shen; Yiqian Zhu; Yerem Yeghiazarians; Wei Zhu; Yongmei Chen; Michael T. Lawton; William L. Young; Guo-Yuan Yang

Circulating blood endothelial progenitor cells (EPCs) contribute to postnatal vasculogenesis, providing a novel therapeutic target for vascular diseases. However, the molecular mechanism of EPC-induced vasculogenesis is unknown. Interleukin-6 plays multiple functions in angiogenesis and vascular remodeling. Our previous study demonstrated that the polymorphism (174G > C) in IL-6 gene promoter was associated with brain vascular disease. In this study, we investigated if IL-6 receptor is expressed in human EPCs derived from circulating mononuclear cells, and if interleukin-6 (IL-6) stimulates EPC angiogenesis in vitro. First, we isolated and cultured mononuclear cells from adult human circulating blood. We obtained EPC clones that were further cultured and expended for the angiogenesis study. We found that the EPCs possessed human mature endothelial cell phenotypes; however, they proliferated much faster than mature endothelial cells (P <0.05). We then found that IL-6 receptor (gp-80) was expressed in the EPCs, and that administration of IL-6 could activate receptor gp80/gp130 signaling pathways including downstream extracellular signal-regulated kinase 1/2 and STAT3 phosphorylation in EPCs. Furthermore, IL-6 stimulated EPC proliferation, migration, and matrigel tube formation in a dose-dependent manner (P <0.05); anti-IL-6 antibodies or IL-6 receptor could abolish these effects (P <0.05). These results suggest that IL-6 plays a crucial role in the biologic behavior of blood-derived EPCs, which may help clarify the mechanism of IL-6 inflammatory-related diseases.


Annals of Neurology | 2006

Interleukin-6 involvement in brain arteriovenous malformations.

Yongmei Chen; Ludmila Pawlikowska; Jianhua S. Yao; Fanxia Shen; Wenwu Zhai; Achal S. Achrol; Michael T. Lawton; Pui-Yan Kwok; Guo-Yuan Yang; William L. Young

We recently reported that the GG genotype of the interleukin‐6 (IL‐6)–174G>C promoter polymorphism is associated with clinical presentation of intracranial hemorrhage in brain arteriovenous malformation (AVM) patients. In this study, we investigated whether tissue IL‐6 expression was associated with IL‐6–174G>C genotype, and whether IL‐6 was linked to downstream targets involved in angiogenesis and vascular instability. Our results showed that the highest IL‐6 protein levels in brain AVM tissue were associated with IL‐6–174GG genotype (GG: 57.7 ± 20.2; GC: 35.6 ± 26.6; CC: 13.9 ± 10.2pg/mg; p = 0.001). IL‐6 protein levels were increased in AVM tissue from patients with hemorrhagic presentation compared with patients without hemorrhage (55 ± 22 vs 40 ± 27pg/mg; p = 0.038). IL‐6 messenger RNA expression strongly correlated with messenger RNA levels of IL‐1β, tumor necrosis factor‐α, IL‐8, matrix metalloproteinase‐3 (MMP‐3), MMP‐9, and MMP‐12. We further investigated the plausibility of IL‐6 being an upstream cytokine responsible for initiating the angiogenic cascade by cell culture and animal experiments. IL‐6 induced MMP‐3 and MMP‐9 expression and activity in mouse brain and increased proliferation and migration of cerebral endothelial cells. Together, our results suggest that the IL‐6 genotype associated with intracranial hemorrhage modulates IL‐6 expression in brain AVM tissue, which is consistent with the hypothesis that inflammatory processes induce angiogenic activity possibly contributory to brain AVM intracranial hemorrhage. Ann Neurol 2005


Stroke | 2005

Angiopoietin-2 Facilitates Vascular Endothelial Growth Factor-Induced Angiogenesis in the Mature Mouse Brain

Yiqian Zhu; Chanhung Lee; Fanxia Shen; Rose Du; William L. Young; Guo-Yuan Yang

Background and Purpose— A better understanding of angiogenic factors and their effects on cerebral angiogenesis is necessary for the development of effective therapeutic strategies for ischemic brain injury. Vascular endothelial growth factor (VEGF) has been shown to induce angiogenesis in the adult mouse brain. However, the function of angiopoietin-2 (Ang-2) in cerebral angiogenesis has not been clarified. The goal of this study was to identify the combined effects of VEGF and Ang-2 on cerebral angiogenesis and the blood–brain barrier (BBB). Methods— Six groups of 6 adult male CD-1 mice underwent AdlacZ (viral vector control), AdVEGF, AdAng2, VEGF protein, VEGF protein plus AdAng2, or saline (negative control) injection. Microvessels were counted using lectin staining on tissue sections after 2 weeks of treatment. Matrix metalloproteinase-9 (MMP-9) activity was determined by zymography. The presence of zonula occludens-1 (ZO-1) protein was determined by Western blot and immunohistochemistry. Results— Mice treated with VEGF protein infusion plus AdAng-2 significantly increased microvessel counts relative to all other groups (P<0.05). The changes in MMP-9 activity paralleled the reduced ZO-1 expression in the VEGF plus Ang-2–treated group compared with the other 5 groups (P<0.05). Double-labeled immunostaining demonstrated that ZO-1–positive staining was significantly decreased on the microvessel wall in the VEGF plus Ang-2–treated group. Conclusions— Our study demonstrates that the combination of VEGF and Ang-2 promotes more angiogenesis compared with VEGF alone. Furthermore, the combination of VEGF and Ang-2 may lead to BBB disruption because it increases MMP-9 activity and inhibits ZO-1 expression.


Stroke | 2006

Adeno-associated viral-vector-mediated hypoxia-inducible vascular endothelial growth factor gene expression attenuates ischemic brain injury after focal cerebral ischemia in mice.

Fanxia Shen; Hua Su; Yongfeng Fan; Yongmei Chen; Yiqian Zhu; Weizhong Liu; William L. Young; Guo-Yuan Yang

Background and Purpose— Exogenous delivery of vascular endothelial growth factor gene (VEGF) may provide a useful approach to the treatment of brain ischemia. We investigated the use of a hypoxia-responsive element to control VEGF expression given for neuroprotection. Methods— Three groups (n=36) of mice received AAVH9-VEGF, AAVH9-lacZ, or saline injection. Five days after gene transfer, the mice underwent 45 minutes of transient middle cerebral artery occlusion (tMCAO) followed by 1 to 7 days of reperfusion. Infarct volume was determined using cresyl violet staining; neuronal injury was examined using TUNEL, cleaved caspase-3, and fluoro-Jade B staining. Results— Hypoxia-inducible factor-1 (HIF-1) was overexpressed after tMCAO in the ischemic hemisphere in the brain. Expression of lacZ, mediated by AAV-lacZ, was seen before and after tMCAO; however, AAVH9-lacZ-mediated lacZ expression was detected only after tMCAO. Infarct volume was smaller in the AAVH9-VEGF-transduced group compared with AAVH9-lacZ and saline groups (55% reduction, P<0.05) with reduced TUNEL (29±5% and 30±7% versus 12±3%, P<0.05), cleaved caspase-3 (20±3% and 21±5% versus 13±4%, P<0.05) and fluoro-Jade B (23±3% and 24±5% versus 12±5%, P<0.05) -positive neurons, respectively. Conclusion— Exogenous expression of VEGF through AAVH9-VEGF gene transfer 5 days before the onset of ischemia provides neuroprotection. Hypoxia-responsive element is a viable strategy of restricting VEGF expression to areas of ischemia to minimize adverse effects of therapy on adjacent normal parenchyma.


Annals of Neurology | 2011

Arteriovenous malformation in the adult mouse brain resembling the human disease

Espen J. Walker; Hua Su; Fanxia Shen; Eun-Jung Choi; S. Paul Oh; Grant Chen; Michael T. Lawton; Helen Kim; Yongmei Chen; Wanqiu Chen; William L. Young

Brain arteriovenous malformations (bAVMs) are an important cause of hemorrhagic stroke. The underlying mechanisms are not clear. No animal model for adult bAVM is available for mechanistic exploration. Patients with hereditary hemorrhagic telangiectasia type 2 (HHT2) with activin receptor‐like kinase 1 (ALK1; ACVRL1) mutations have a higher incidence of bAVM than the general population. We tested the hypothesis that vascular endothelial growth factor (VEGF) stimulation with regional homozygous deletion of Alk1 induces severe dysplasia in the adult mouse brain, akin to human bAVM.


Neurosurgery | 2006

Expression of hypoxia-inducible factor-1 and vascular endothelial growth factor in response to venous hypertension.

Yiqian Zhu; Michael T. Lawton; Rose Du; Yamin Shwe; Yongmei Chen; Fanxia Shen; William L. Young; Guo-Yuan Yang

OBJECTIVE:Experimentally, a fistula created surgically between the carotid artery and jugular vein, together with occlusion of venous sinuses, generate venous hypertension, which can induce dural arteriovenous fistula formation intracranially in rats. Our aim was to study the effect of nonischemic venous hypertension on the elaboration of the angiogenic signal, hypoxia-inducible factor-1 (HIF-1), and its downstream signal, vascular endothelial growth factor (VEGF). METHODS:Sixty rats were exposed to venous hypertension for periods ranging from 4 hours to 3 weeks. Western blot analysis, transbinding assays, enzyme-linked immunosorbent assays, and immunohistochemistry quantified HIF-1 and VEGF expression in brain. Forty-eight control rats underwent similar surgical procedures without creating venous hypertension. Cerebral blood flow was measured at baseline, after surgery, and before sacrifice. RESULTS:Venous hypertension did not impair cerebral blood flow. Relative to controls, HIF-1 expression increased fivefold in response to venous hypertension (P < 0.005), with peak expression 1 day later localized to endothelial cells in venules next to the sagittal sinus. VEGF expression also increased threefold in response to venous hypertension (P < 0.05), with peak expression 7 days later localized to parasagittal astrocytes. HIF-1 and VEGF were minimally expressed in rat normal venous pressures. CONCLUSION:In this model, venous hypertension stimulates angiogenesis by a mechanism other than ischemia. HIF-1 expression may result from dilation of parasagittal veins and endothelial deformation. HIF-1 and VEGF seem to be molecular agents that convert venous hypertension into intracellular signals and angiogenesis activity.


Stroke | 2012

Bevacizumab Attenuates VEGF-Induced Angiogenesis and Vascular Malformations in the Adult Mouse Brain

Espen J. Walker; Hua Su; Fanxia Shen; Vincent Degos; Gregory Amend; Kristine Jun; William L. Young

Background and Purpose— Vascular endothelial growth factor (VEGF) expression is elevated in human brain arteriovenous malformations (bAVM). We have developed a bAVM model in the adult mouse by focal Alk1 gene deletion and human VEGF stimulation. We hypothesized that once the abnormal vasculature has been established, tonic VEGF stimulation is necessary to maintain the abnormal phenotype, and VEGF antagonism by bevacizumab (Avastin) would reduce vessel density and attenuate the dysplastic vascular phenotype. Methods— Angiogenesis and bAVM were induced by injection of adeno-associated viral vector expressing human VEGF alone into the brain of wild-type mice or with adenoviral vector expressing Cre recombinase (Ad-Cre) into Alk12f/2f mice. Six weeks later, bevacizumab or trastuzumab (Herceptin, bevacizumab control) was administered. Vessel density, dysplasia index, vascular cell proliferation and apoptosis, and human IgG were assessed (n=6/group). Results— Compared with trastuzumab (15 mg/kg), administration of 5, 10, and 15 mg/kg of bevacizumab to adeno-associated viral vector expressing human VEGF treated wild-type mice reduced focal vessel density (P<0.05); administration of 5 mg/kg bevacizumab decreased proliferating vascular cells (P=0.04) and increased TUNEL-positive vascular cells (P=0.03). More importantly, bevacizumab (5 mg/kg) treatment reduced both vessel density (P=0.01) and dysplasia index (P=0.02) in our bAVM model. Human IgG was detected in the vessel wall and in the parenchyma in the angiogenic foci of bevacizumab-treated mice. Conclusions— We provide proof-of-principle that, once abnormal AVM vessels have formed, VEGF antagonism may reduce the number of dysplastic vessels and should be evaluated further as a therapeutic strategy for the human disease.


Journal of Cerebral Blood Flow and Metabolism | 2008

Overexpression of Netrin-1 Induces Neovascularization in the Adult Mouse Brain:

Yongfeng Fan; Fanxia Shen; Yongmei Chen; Qi Hao; Weizhong Liu; Hua Su; William L. Young; Guo-Yuan Yang

Netrin-1 is a critical molecule for axonal pathfinding during embryo development, and because of its structural homology to the endothelial mitogens, it may share its effects on vascular network formation. Using an adeno-associated viral netrin-1 vector (AAV-NT-1) gene transfer, we demonstrated that netrin-1 was able to stimulate the proliferation and migration of human cerebral endothelial cells (HCECs) and human aortic smooth muscle cells (HASMCs) compared with the control (P < 0.05), and could also promote HCEC tube formation on matrigel (P < 0.05) in vitro. Moreover, netrin-1 hyperstimulation could promote focal neovascularization (P < 0.05) in the adult brain in vivo. Unlike VEGF-induced microvessel increase, netrin-1-induced newly formed vessels showed an artery-like phenotype, with an intact endothelial cell monolayer surrounded by multiple cell layers, including smooth muscle cells and an astrocyte-connected outer layer. Our findings suggest that netrin-1 plays an important role in promoting blood vessel formation in the adult rodent central nervous system, and could have broad implication in cerebrovascular development and remodeling.

Collaboration


Dive into the Fanxia Shen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hua Su

University of California

View shared research outputs
Top Co-Authors

Avatar

Guo-Yuan Yang

University of California

View shared research outputs
Top Co-Authors

Avatar

Yongmei Chen

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael T. Lawton

Barrow Neurological Institute

View shared research outputs
Top Co-Authors

Avatar

Yiqian Zhu

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qi Hao

University of California

View shared research outputs
Top Co-Authors

Avatar

Yongfeng Fan

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge