2,038
Views
24
CrossRef citations to date
0
Altmetric
Research Article

Efficient treatment of Parkinson’s disease using ultrasonography-guided rhFGF20 proteoliposomes

, , , , , , , , , & show all
Pages 1560-1569 | Received 17 Apr 2018, Accepted 28 May 2018, Published online: 25 Jul 2018

References

  • Aryal M, Arvanitis CD, Alexander PM, et al. (2014). Ultrasound-mediated blood-brain barrier disruption for targeted drug delivery in the central nervous system. Adv Drug Deliv Rev 72:94–109.
  • Aryal M, Vykhodtseva N, Zhang Y-Z, et al. (2015). Multiple sessions of liposomal doxorubicin delivery via focused ultrasound mediated blood-brain barrier disruption: a safety study. J Control Release 204:60–9.
  • Basu A, Li X, Leong SS. (2011). Refolding of proteins from inclusion bodies: rational design and recipes. Appl Microbiol Biotechnol 92:241–51.
  • Beenken A, Mohammadi M. (2009). The FGF family: biology, pathophysiology and therapy. Nat Rev Drug Discov 8:235–53.
  • Correia AS. (2007). Fibroblast growth factor-20 increases the yield of midbrain dopaminergic neurons derived from human embryonic stem cells. Front Neuroanat 1:4.
  • Grothe C, Timmer M, Scholz T, et al. (2004). Fibroblast growth factor-20 promotes the differentiation of Nurr1-overexpressing neural stem cells into tyrosine hydroxylase-positive neurons. Neurobiol Dis 17:163–70.
  • Hartmann A, Hunot S, Michel PP, et al. (2000). Caspase-3: a vulnerability factor and final effector in apoptotic death of dopaminergic neurons in Parkinson’s disease. Proc Natl Acad Sci USA 97:2875–80.
  • Itoh N, Ohta H. (2013). Roles of FGF20 in dopaminergic neurons and Parkinson’s disease. Front Mol Neurosci 6:15.
  • Jia X, Tian H, Tang L, et al. (2015). High-efficiency expression of TAT-bFGF fusion protein in Escherichia coli and the effect on hypertrophic scar tissue. PLoS One 10:e0117448.
  • Kobus T, Zervantonakis IK, Zhang Y, et al. (2016). Growth inhibition in a brain metastasis model by antibody delivery using focused ultrasound-mediated blood-brain barrier disruption. J Control Release 238:281–8.
  • Konofagou EE. (2012). Optimization of the ultrasound-induced blood-brain barrier opening. Theranostics 2:1223–37.
  • Liu X, Chen Y, Wu X, et al. (2012). SUMO fusion system facilitates soluble expression and high production of bioactive human fibroblast growth factor 23 (FGF23). Appl Microbiol Biotechnol 96:103–11.
  • Middelberg AP. (2002). Preparative protein refolding. Trends Biotechnol 20:437–43.
  • O’Reilly MA, Waspe AC, Ganguly M, et al. (2011). Focused-ultrasound disruption of the blood-brain barrier using closely-timed short pulses: influence of sonication parameters and injection rate. Ultrasound Med Biol 37:587–94.
  • Ohmachi S, Mikami T, Konishi M, et al. (2003). Preferential neurotrophic activity of fibroblast growth factor-20 for dopaminergic neurons through fibroblast growth factor receptor-1c. J Neurosci Res 72:436–43.
  • Ohmachi S, Watanabe Y, Mikami T, et al. (2000). FGF-20, a novel neurotrophic factor, preferentially expressed in the substantia nigra pars compacta of rat brain. Biochem Biophys Res Commun 277:355–60.
  • Pan J, Li H, Wang Y, et al. (2012). Fibroblast growth factor 20 (FGF20) polymorphism is a risk factor for Parkinson’s disease in Chinese population. Parkinsonism Relat Disord 18:629–31.
  • Park E-J, Zhang Y-Z, Vykhodtseva N, et al. (2012). Ultrasound-mediated blood-brain/blood-tumor barrier disruption improves outcomes with trastuzumab in a breast cancer brain metastasis model. J Control Release 163:277–84.
  • Patel MM, Patel BM. (2017). Crossing the blood-brain barrier: recent advances in drug delivery to the brain. CNS Drugs 31:109–33.
  • Schober A. (2004). Classic toxin-induced animal models of Parkinson's disease: 6-OHDA and MPTP. Cell Tissue Res 318:215–24.
  • Shimada H, Yoshimura N, Tsuji A, et al. (2009). Differentiation of dopaminergic neurons from human embryonic stem cells: modulation of differentiation by FGF-20. J Biosci Bioeng 107:447–54.
  • Sleeman IJ, Boshoff EL, Duty S. (2012). Fibroblast growth factor-20 protects against dopamine neuron loss in vitro and provides functional protection in the 6-hydroxydopamine-lesioned rat model of Parkinson’s disease. Neuropharmacology 63:1268–77.
  • Sun XW, Wang XH, Yao YB. (2014). Co-expression of Dsb proteins enables soluble expression of a single-chain variable fragment (scFv) against human type 1 insulin-like growth factor receptor (IGF-1R) in E. coli. World J Microbiol Biotechnol 30:3221–7.
  • Tatton WG, Chalmers-Redman R, Brown D, et al. (2003). Apoptosis in Parkinson's disease: signals for neuronal degradation. Ann Neurol 53(Suppl. 3):S61–S70,discussion S70–2.
  • Thisse B, Thisse C. (2005). Functions and regulations of fibroblast growth factor signaling during embryonic development. Dev Biol 287:390–402.
  • Thuret S, Bhatt L, O'Leary DDM, et al. (2004). Identification and developmental analysis of genes expressed by dopaminergic neurons of the substantia nigra pars compacta. Mol Cell Neurosci 25:394–405.
  • Tian X-Q, Ni X-W, Xu H-L, et al. (2017). Prevention of doxorubicin-induced cardiomyopathy using targeted MaFGF mediated by nanoparticles combined with ultrasound-targeted MB destruction. Int J Nanomed 12:7103–19.
  • Tian H, Zhao Y, Chen N, et al. (2016). High production in E. coli of biologically active recombinant human fibroblast growth factor 20 and its neuroprotective effects. Appl Microbiol Biotechnol 100:3023–34.
  • Tolleson CM, Fang JY. (2013). Advances in the mechanisms of Parkinson's disease. Discov Med 15:61–6.
  • Vlachos F, Tung YS, Konofagou EE. (2010). Permeability assessment of the focused ultrasound-induced blood-brain barrier opening using dynamic contrast-enhanced MRI. Phys Med Biol 55:5451–66.
  • Walkinshaw G, Waters CM. (1994). Neurotoxin-induced cell death in neuronal PC12 cells is mediated by induction of apoptosis. Neuroscience 63:975–87.
  • Wang H, Xiao Y, Fu L, et al. (2010). High-level expression and purification of soluble recombinant FGF21 protein by SUMO fusion in Escherichia coli. BMC Biotechnol 10:14.
  • Zhu G, Chen G, Shi L, et al. (2015). PEGylated rhFGF-2 conveys long-term neuroprotection and improves neuronal function in a rat model of Parkinson’s disease. Mol Neurobiol 51:32–42.