Radial Extracorporeal Shock Wave Therapy Enhances the Proliferation and Differentiation of Neural Stem Cells by Notch, PI3K/AKT, and Wnt/β-catenin Signaling

Jing Zhang, Nan Kang, Xiaotong Yu, Yuewen Ma, Xining Pang, Jing Zhang, Nan Kang, Xiaotong Yu, Yuewen Ma, Xining Pang

Abstract

Neural stem cell (NSC) proliferation and differentiation play a pivotal role in the repair of brain function in central nervous system (CNS) diseases. Radial extracorporeal shock wave therapy (rESWT) is a non-invasive and innovative treatment for many conditions, yet little is known about the effects of this treatment on NSCs. Mouse NSCs (NE-4C) were exposed to rESWT with 1.0, 1.5, 2.0, 2.5, 3.0, and 3.5 bar (500 impulses, and 2 Hz) in vitro. Cell viability test results indicated that rESWT, at a dose of 2.5 bar, 500 impulses, and 2 Hz, increased NE-4C viability within 72 h, and that the PI3K/AKT pathway was involved in its mechanisms. Exposure to rESWT also affected proliferation and differentiation of NE-4C after 8 weeks, which may be associated with Wnt/β-catenin and Notch pathways. This assessment is corroborated by the ability of inhibitors of Wnt/β-catenin [Dickkopf-1 (Dkk-1)] and the Notch pathway (DAPT) to weaken proliferation and differentiation of NSCs. In summary, a proper dose of rESWT enhanced NSCs augment via the PI3K/AKT pathway initially. Also, Wnt/β-catenin and the Notch pathway play important roles in regulation of the long-term efficacy of rESWT. This study reveals a novel approach to culture NSCs in vitro and support neurogenesis.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
The effect of different doses of rESWT applied on the cultures. The multiplication of NE-4C cells was analyzed using an MTT cell proliferation assay (Promega, Japan). rESWT at a dose of 2.5 bar, 500 impulses, 2 Hz significantly stimulated the number of NE-4C cells at 72 h compared with the control group. Bar: 20 μm. (A) Different doses of rESWT ranging from 1.0 to 3.5 bar were used in cultures. The proliferation of NE-4C cells was tested at 12, 24, 36, 48, 60, and 72 h after rESWT. Below the 2.5 bar, 500 impulses, 2 Hz dose, rESWT increased proliferation in a quantity-dependent manner. Over the 2.5 bar, 500 impulses, 2 Hz dose, rESWT had an opposite effect (B).
Figure 2
Figure 2
NSE expression. NSE expression was assessed by immunofluorescence, Western blot analysis, and qRT-PCR. Fluorescence signals were analyzed by an Axiovert 200 inverted microscope. Nuclei were stained with DAPI. Bar: 20 μm (A–C). The Western blot shows a visible difference in three groups (D). Data are reported as the mean ± SD of significant difference from the control group at both week 8 (*P < 0.05) and week 12 (**P < 0.01) and from the Dkk-1 inhibitor group (***P < 0.001) (E). NSE mRNA expression was improved at 8 and 12 weeks compared with the control group (**P < 0.01) and Dkk-1 inhibitor group (***P < 0.001) (F).
Figure 3
Figure 3
β-tubulin III expression. β-Tubulin III expression was improved at 8 and 12 weeks compared with the control group. The Dkk-1 inhibitor suppressed β-tubulin III expression with regard to both the protein (A–E) and mRNA (F) levels as determined by immunofluorescence staining (A–C), Western blot (D,E), and qRT-PCR (F) analyses. β-Tubulin III fluorescence is shown in green and nuclei staining is shown in blue. Bar: 20 μm (**p < 0.01, ***P < 0.001).
Figure 4
Figure 4
Western blot and Real-time PCR findings of PI3K and AKT. The Western blot results demonstrate the enhanced expression of PI3K and AKT at 24, 48, and 72 h after rESWT in two independent experiments (***P A,B,D,E). PI3K and AKT mRNA expression, as measured with qRT-PCR, are also increased at 24, 48, and 72 h using rESWT (***P < 0.001) (in C,F).
Figure 5
Figure 5
Wnt3a expression. Wnt3a expression was determined at 1, 4, 8, and 12 weeks. Immunofluorescence microscopic findings of Wnt3a (green color) show that the expression increased compared with the control group and decreased in the Dkk-1 inhibitor group (A–C). Western blot findings demonstrate that the prominent improvement in expression was revealed only at 8 and 12 weeks relative to the control group (D,E). The same results are seen for the mRNA expression (F). (*P < 0.05, **P < 0.01, ***P < 0.001). The data are presented as the mean ± SD.
Figure 6
Figure 6
β-catenin expression. β-catenin (green color) immunofluorescence findings (A–C), and Western blot findings (D,E) demonstrate the augmented expression at weeks 8 and 12 compared with the control group, and reduced expression induced by the Dkk-1 inhibitor (***P < 0.001). β-catenin mRNA expression was also improved relative to control group at 8 weeks (**P < 0.01) and 12 weeks (***P < 0.001) and reduced in the Dkk-1 inhibitor group at 8 weeks (*P < 0.05) and 12 weeks (***P < 0.001) (F). Data are reported as the mean ± SD.
Figure 7
Figure 7
LRP-6, Axin, Frizzled, and GSK3β expression. qRT-PCR was used to test the mRNA expression of LRP-6, Axin, Frizzled, and GSK3β. Results revealed distinct differences between three groups on weeks 8 and 12. LRP-6, Axin, and Frizzled mRNA expression increased after rESWT and was suppressed by the Dkk-1 inhibitor (A–C). GSK3β expression was opposite to that seen for LRP-6, Axin, and Frizzled mRNA (D). (*P < 0.05, **P < 0.01, ***P < 0.001).
Figure 8
Figure 8
Notch1 signaling, nestin, and NSE expression after rESWT. Notch1, NICD, and Jagged1 expression was enhanced after rESWT on weeks 8 and 12 (**P E,G,I). mRNA expression for Notch1, Jagged1, and Hes1 was augmented compared with the control group and DAPT group (**P < 0.01, ***P < 0.001) (F,H,J). Notch1 signaling was upregulated and expression of NSE, nestin, and mRNA was increased and was inhibited by DAPT on weeks 8 and 12 (***P < 0.001, **P < 0.01) (A–D).

References

    1. Ottoboni L, Merlini A, Martino G. Neural Stem Cell Plasticity: Advantages in Therapy for the Injured Central Nervous System. Frontiers in cell and developmental biology. 2017;5:52. doi: 10.3389/fcell.2017.00052.
    1. Zhang J, Jiao J. Molecular Biomarkers for Embryonic and Adult Neural Stem Cell and Neurogenesis. BioMed research international. 2015;2015:727542.
    1. Arvidsson A, Collin T, Kirik D, Kokaia Z, Lindvall O. Neuronal replacement from endogenous precursors in the adult brain after stroke. Nature medicine. 2002;8:963–970. doi: 10.1038/nm747.
    1. Topchiy E, et al. Nox4-generated superoxide drives angiotensin II-induced neural stem cell proliferation. Developmental neuroscience. 2013;35:293–305. doi: 10.1159/000350502.
    1. Cernilogar FM, Di Giaimo R, Rehfeld F, Cappello S, Lie DC. RNA interference machinery-mediated gene regulation in mouse adult neural stem cells. BMC neuroscience. 2015;16:60. doi: 10.1186/s12868-015-0198-7.
    1. Nakagomi N, et al. Endothelial cells support survival, proliferation, and neuronal differentiation of transplanted adult ischemia-induced neural stem/progenitor cells after cerebral infarction. Stem cells. 2009;27:2185–2195. doi: 10.1002/stem.161.
    1. von Bohlen Und Halbach O. Immunohistological markers for staging neurogenesis in adult hippocampus. Cell and tissue research. 2007;329:409–420. doi: 10.1007/s00441-007-0432-4.
    1. Bergstrom T, Forsberg-Nilsson K. Neural stem cells: brain building blocks and beyond. Upsala journal of medical sciences. 2012;117:132–142. doi: 10.3109/03009734.2012.665096.
    1. Shi X, et al. miR-381 Regulates Neural Stem Cell Proliferation and Differentiation via Regulating Hes1 Expression. PloS one. 2015;10:e0138973. doi: 10.1371/journal.pone.0138973.
    1. Ma J, et al. Neural stem cell transplantation promotes behavioral recovery in a photothrombosis stroke model. International journal of clinical and experimental pathology. 2015;8:7838–7848.
    1. Ojeda L, et al. Critical role of PI3K/Akt/GSK3beta in motoneuron specification from human neural stem cells in response to FGF2 and EGF. PloS one. 2011;6:e23414. doi: 10.1371/journal.pone.0023414.
    1. Adachi K, et al. Beta-catenin signaling promotes proliferation of progenitor cells in the adult mouse subventricular zone. Stem cells. 2007;25:2827–2836. doi: 10.1634/stemcells.2007-0177.
    1. Edri R, et al. Analysing human neural stem cell ontogeny by consecutive isolation of Notch active neural progenitors. Nature communications. 2015;6:6500. doi: 10.1038/ncomms7500.
    1. Le Belle JE, et al. Proliferative neural stem cells have high endogenous ROS levels that regulate self-renewal and neurogenesis in a PI3K/Akt-dependant manner. Cell stem cell. 2011;8:59–71. doi: 10.1016/j.stem.2010.11.028.
    1. Bowman AN, van Amerongen R, Palmer TD, Nusse R. Lineage tracing with Axin2 reveals distinct developmental and adult populations of Wnt/beta-catenin-responsive neural stem cells. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:7324–7329. doi: 10.1073/pnas.1305411110.
    1. Lie DC, et al. Wnt signalling regulates adult hippocampal neurogenesis. Nature. 2005;437:1370–1375. doi: 10.1038/nature04108.
    1. Haack F, Lemcke H, Ewald R, Rharass T, Uhrmacher AM. Spatio-temporal model of endogenous ROS and raft-dependent WNT/beta-catenin signaling driving cell fate commitment in human neural progenitor cells. PLoS computational biology. 2015;11:e1004106. doi: 10.1371/journal.pcbi.1004106.
    1. Falk R, et al. Generation of anti-Notch antibodies and their application in blocking Notch signalling in neural stem cells. Methods. 2012;58:69–78. doi: 10.1016/j.ymeth.2012.07.008.
    1. Yoon K, Gaiano N. Notch signaling in the mammalian central nervous system: insights from mouse mutants. Nature neuroscience. 2005;8:709–715. doi: 10.1038/nn1475.
    1. Li Y, Tzatzalos E, Kwan KY, Grumet M, Cai L. Transcriptional Regulation of Notch1 Expression by Nkx6.1 in Neural Stem/Progenitor Cells during Ventral Spinal CordDevelopment . Scientific reports. 2016;6:38665. doi: 10.1038/srep38665.
    1. Stump G, et al. Notch1 and its ligands Delta-like and Jagged are expressed and active in distinct cell populations in the postnatal mouse brain. Mechanisms of development. 2002;114:153–159. doi: 10.1016/S0925-4773(02)00043-6.
    1. Breunig JJ, Silbereis J, Vaccarino FM, Sestan N, Rakic P. Notch regulates cell fate and dendrite morphology of newborn neurons in the postnatal dentate gyrus. Proceedings of the National Academy of Sciences of the United States of America. 2007;104:20558–20563. doi: 10.1073/pnas.0710156104.
    1. MC DA, et al. Extracorporeal shockwaves as regenerative therapy in orthopedic traumatology: a narrative review from basic research to clinical practice. Journal of biological regulators and homeostatic agents. 2016;30:323–332.
    1. Schmitz C, et al. Efficacy and safety of extracorporeal shock wave therapy for orthopedic conditions: a systematic review on studies listed in the PEDro database. British medical bulletin. 2015;116:115–138.
    1. Aschermann I, et al. Extracorporal Shock Waves Activate Migration, Proliferation and Inflammatory Pathways in Fibroblasts and Keratinocytes, and Improve Wound Healing in an Open-Label, Single-Arm Study in Patients with Therapy-Refractory Chronic Leg Ulcers. Cellular physiology and biochemistry: international journal of experimental cellular physiology, biochemistry, and pharmacology. 2017;41:890–906. doi: 10.1159/000460503.
    1. Lee JH, Cho SH. Effect of extracorporeal shock wave therapy on denervation atrophy and function caused by sciatic nerve injury. Journal of physical therapy science. 2013;25:1067–1069. doi: 10.1589/jpts.25.1067.
    1. Yuen CM, et al. Extracorporeal shock wave effectively attenuates brain infarct volume and improves neurological function in rat after acute ischemic stroke. American journal of translational research. 2015;7:976–994.
    1. Kang N, Zhang J, Yu X, Ma Y. Radial extracorporeal shock wave therapy improves cerebral blood flow and neurological function in a rat model of cerebral ischemia. American journal of translational research. 2017;9:2000–2012.
    1. Leone L, et al. Extracorporeal Shock Wave Treatment (ESWT) enhances the in vitro-induced differentiation of human tendon-derived stem/progenitor cells (hTSPCs) Oncotarget. 2016;7:6410–6423. doi: 10.18632/oncotarget.7064.
    1. Hochstrasser T, Frank HG, Schmitz C. Dose-dependent and cell type-specific cell death and proliferation following in vitro exposure to radial extracorporeal shock waves. Scientific reports. 2016;6:30637. doi: 10.1038/srep30637.
    1. Gollwitzer H, et al. Radial extracorporeal shock wave therapy (rESWT) induces new bone formation in vivo: results of an animal study in rabbits. Ultrasound in medicine & biology. 2013;39:126–133. doi: 10.1016/j.ultrasmedbio.2012.08.026.
    1. Mangone G, Veliaj A, Postiglione M, Viliani T, Pasquetti P. Radial extracorporeal shock-wave therapy in rotator cuff calcific tendinosis. Clinical cases in mineral and bone metabolism: the official journal of the Italian Society of Osteoporosis, Mineral Metabolism, and Skeletal Diseases. 2010;7:91–96.
    1. Storz Medical Masterpuls® MP200. Indications-concise instruction,
    1. Schuh CM, et al. In vitro extracorporeal shock wave treatment enhances stemness and preserves multipotency of rat and human adipose-derived stem cells. Cytotherapy. 2014;16:1666–1678. doi: 10.1016/j.jcyt.2014.07.005.
    1. Raabe O, et al. Effect of extracorporeal shock wave on proliferation and differentiation of equine adipose tissue-derived mesenchymal stem cells in vitro. American journal of stem cells. 2013;2:62–73.
    1. Chan WS, et al. Differential regulation of proliferation and neuronal differentiation in adult rat spinal cord neural stem/progenitors by ERK1/2, Akt, and PLCgamma. Frontiers in molecular neuroscience. 2013;6:23. doi: 10.3389/fnmol.2013.00023.
    1. Zhang YH, et al. The PI3K-AKT-mTOR pathway activates recovery from general anesthesia. Oncotarget. 2016;7:40939–40952. doi: 10.18632/oncotarget.10172.
    1. Wu Y, et al. CXCL12 increases human neural progenitor cell proliferation through Akt-1/FOXO3a signaling pathway. Journal of neurochemistry. 2009;109:1157–1167. doi: 10.1111/j.1471-4159.2009.06043.x.
    1. Jiang LL, et al. miR-93 Promotes Cell Proliferation in Gliomas through Activation of PI3K/Akt Signaling Pathway. Oncotarget. 2015;6:8286–8299. doi: 10.18632/oncotarget.3221.
    1. Hirabayashi Y, et al. The Wnt/beta-catenin pathway directs neuronal differentiation of cortical neural precursor cells. Development. 2004;131:2791–2801. doi: 10.1242/dev.01165.
    1. Dunty WC, et al. Transcriptional profiling of Wnt3a mutants identifies Sp transcription factors as essential effectors of the Wnt/beta-catenin pathway in neuromesodermal stem cells. PloS one. 2014;9:e87018. doi: 10.1371/journal.pone.0087018.
    1. McCubrey JA, et al. Effects of mutations in Wnt/beta-catenin, hedgehog, Notch and PI3K pathways on GSK-3 activity-Diverse effects on cell growth, metabolism and cancer. Biochimica et biophysica acta. 2016;1863:2942–2976. doi: 10.1016/j.bbamcr.2016.09.004.
    1. Ling L, Nurcombe V, Cool SM. Wnt signaling controls the fate of mesenchymal stem cells. Gene. 2009;433:1–7. doi: 10.1016/j.gene.2008.12.008.
    1. Zhang X, et al. Macrophage migration inhibitory factor promotes proliferation and neuronal differentiation of neural stem/precursor cells through Wnt/beta-catenin signal pathway. International journal of biological sciences. 2013;9:1108–1120. doi: 10.7150/ijbs.7232.
    1. Niehrs C. Function and biological roles of the Dickkopf family of Wnt modulators. Oncogene. 2006;25:7469–7481. doi: 10.1038/sj.onc.1210054.
    1. Kong JH, et al. Notch activity modulates the responsiveness of neural progenitors to sonic hedgehog signaling. Developmental cell. 2015;33:373–387. doi: 10.1016/j.devcel.2015.03.005.
    1. Li Y, Tzatzalos E, Kwan KY, Grumet M, Cai L. Transcriptional Regulation of Notch1 Expression by Nkx6.1 in Neural Stem/Progenitor Cells during Ventral Spinal CordDevelopment. Scientific reports. 2016;6:38665. doi: 10.1038/srep38665.
    1. Teixeira, F. G. et al. Modulation of the Mesenchymal Stem Cell Secretome Using Computer-Controlled Bioreactors: Impact on NeuronalCell Proliferation, Survival and Differentiation. Scientific reports6, doi:Artn 2779110.1038/Srep27791 (2016).
    1. Trujillo-Paredes N, et al. Regulation of differentiation flux by Notch signalling influences the number of dopaminergic neurons in the adult brain. Biology open. 2016;5:336–347. doi: 10.1242/bio.013383.
    1. Robinson AP, Foraker JE, Ylostalo J, Prockop DJ. Human stem/progenitor cells from bone marrow enhance glial differentiation of rat neural stem cells: a role for transforming growth factor beta and Notch signaling. Stem cells and development. 2011;20:289–300. doi: 10.1089/scd.2009.0444.
    1. Aguirre A, Rubio ME, Gallo V. Notch and EGFR pathway interaction regulates neural stem cell number and self-renewal. Nature. 2010;467:323–327. doi: 10.1038/nature09347.
    1. Liu Z, Sneve M, Haroldson TA, Smith JP, Drewes LR. Regulation of Monocarboxylic Acid Transporter 1 Trafficking by the Canonical Wnt/beta-Catenin Pathway in Rat Brain Endothelial Cells Requires Cross-talk with Notch Signaling. The Journal of biological chemistry. 2016;291:8059–8069. doi: 10.1074/jbc.M115.710277.
    1. Kondeti V, et al. Differential regulation of cysteinyl leukotriene receptor signaling by protein kinase C in human mast cells. PloS one. 2013;8:e71536. doi: 10.1371/journal.pone.0071536.
    1. Bustin SA, et al. The MIQE guidelines: minimum information for publication of quantitative real-time PCR experiments. Clinical chemistry. 2009;55:611–622. doi: 10.1373/clinchem.2008.112797.

Source: PubMed

3
Suscribir