Archives

  • 2018-07
  • 2018-10
  • 2018-11
  • 2019-04
  • 2019-05
  • 2019-06
  • 2019-07
  • 2019-08
  • 2019-09
  • 2019-10
  • 2019-11
  • 2019-12
  • 2020-01
  • 2020-02
  • 2020-03
  • 2020-04
  • 2020-05
  • 2020-06
  • 2020-07
  • 2020-08
  • 2020-09
  • 2020-10
  • 2020-11
  • 2020-12
  • 2021-01
  • 2021-02
  • 2021-03
  • 2021-04
  • 2021-05
  • 2021-06
  • 2021-07
  • 2021-08
  • 2021-09
  • 2021-10
  • 2021-11
  • 2021-12
  • 2022-01
  • 2022-02
  • 2022-03
  • 2022-04
  • 2022-05
  • 2022-06
  • 2022-07
  • 2022-08
  • 2022-09
  • 2022-10
  • 2022-11
  • 2022-12
  • 2023-01
  • 2023-02
  • 2023-03
  • 2023-04
  • 2023-05
  • 2023-06
  • 2023-07
  • 2023-08
  • 2023-09
  • 2023-10
  • 2023-11
  • 2023-12
  • 2024-01
  • 2024-02
  • 2024-03
  • 2024-04
  • Phos-tag Biotin BTL-105 Recent research suggests that NSCs t

    2018-11-08

    Recent research suggests that NSCs\' tropism towards neoplastic areas is driven by chemoattractants produced at the tumor site. The intratumoral hypoxic microenvironment activates SDF-1/CXCR4 and VEGF/VEGFR signaling pathways, which may act as downstream effectors to increase NSC tropism (Aboody et al., 2006; Zhao et al., 2008; Ingraham et al., 2011; Rosova et al., 2008; Liu et al., 2010). We found that VEGFR2 expression was critical for the tumor-tropic migratory properties of HB1.F3.CD NSCs. Further analysis showed that the highest VEGFR2 expression was concentrated at focal adhesion complexes and filopodia of migratory cells. This suggests that VEGF-A-mediated cytoskeletal reorganization is present in our studied migratory population. These data are in line with previously published studies that show VEGFA-mediated Phos-tag Biotin BTL-105 reorganization and filopodia formation in migratory endothelial and oligodendrocyte precursor cells (Laquintana et al., 2009; Hayakawa et al., 2011; Rousseau et al., 1997). We further assessed the activation of VEGF-A downstream pathways in our migratory NSCs and their collective role in NSC migration. First, we observed VEGF-A-mediated phosphorylation and activation of VEGF-A\'s binding site, p-VEGFR2 (Tyr1175). Previous publications described Tyr1175 as Phos-tag Biotin BTL-105 being responsible for linking VEGF-A receptor phosphorylation to signal transduction. PLCγ, an upstream kinase that requires Src binding for its activation, was shown to bind the intracellular domain of VEGFR2, leading to activation of the MAP kinase ERK1/2 signaling pathway. PLCγ and ERK1/2 activation lead to podosome formation, cell migration, and proliferation (Lee et al., 2009; Takeuchi et al., 2007). Similarly, our results indicated downstream activation of Src, PLCγ1, and MAP kinase ERK1/2 upon VEGF-A stimulation. We additionally noted high levels of activated p38 in migratory NSCs upon VEGF-A treatment. This finding is consistent with previously published materials that point to VEGF-induced activation of p38 and ERK1/2 as a possible mechanism of focal adhesion and podosome formation in migratory cells (Rousseau et al., 1997). We further observed downstream activation of Akt in migratory cells, achieving its peak 30min after VEGF-A stimulation. Similarly, other publications have demonstrated a VEGF-mediated activation of Akt, a kinase pathway linked to increased migration and inhibition of apoptosis in endothelial cells (Deryugina et al., 2002; Morales-Ruiz et al., 2000). Lastly, we also detected strong FAK activation in migratory NSCs. In agreement with our results, Src was previously shown to bind FAK, leading to increased migration of both oligodendrocyte precursors and endothelial cells. In addition, FAK has been shown to regulate focal adhesion turnover during cell migration and to regulate cell survival (Ilic et al., 2004). Previously published mechanisms of VEGF-mediated downstream pathway activation have been described in endothelial cells, corneal fibroblasts, oligodendrocyte precursor cells, and cancer cells (Olsson et al., 2006; Deryugina et al., 2002; Han et al., 2012; Hayakawa et al., 2011). Our results suggest that VEGF-A-dependent downstream activation of PLCγ1, Akt, and FAK in NSCs may be directly related to cytoskeletal reorganization, podosome formation, and cell migration. This explains our observation of increased VEGFR2 expression in focal adhesion areas of migratory NSCs. This mechanism highlights the upstream signaling pathways that are likely driving the observed differential migratory capacities in NSCs expressing higher levels of VEGFR2. We next studied the role of MMP14 in NSCs\' targeted migration towards diffuse tumor foci. We found that MMP14 was required for NSC migration and that its expression is dependent on VEGFR2 activation in migratory NSCs. Knockdown of VEGFR2 completely blocked MMP14 expression and significantly impaired migration of HB1.F3.CD NSCs. We have thus demonstrated, for the first time, that MMP14 is a possible downstream target of activated VEGFR2 in migratory NSCs. This finding is in agreement with previously published data demonstrating decreased MMP14 activity inhibited the migration of human bone marrow MSCs (Malinowski et al., 2012). The same study also revealed that MMP14 overexpression had the capacity to restore MSC migration. Additional reports observed VEGF-induced MMP activation and extracellular matrix degradation in endothelial cells (Laquintana et al., 2009). Interestingly, several other studies indicated that a reverse pathway was present in malignant cells, including malignant glioma and breast cancer models, where MMP14 was noted to regulate VEGF-A expression through a complex with VEGFR2 and Src, leading to increased xenograft growth and tumor-related angiogenesis (Deryugina et al., 2002). Thus, it is conceivable that in the NSC line the autocrine upregulation of MMP14, mediated by VEGFA/VEGFR2 signaling, is responsible for targeted NSC migration towards malignant areas. Much interest has been placed on various niches that may populate the landscape of malignant gliomas and represent distinct tumor microenvironments, among them hypoxic regions that have been shown to secrete higher levels of VEGF-A and, in turn, attract migratory NSCs. Therefore, it is possible that constant VEGF-A expression may also activate intrinsic VEGFR2 activity in migratory cells in an autocrine manner. This leads to downstream MMP14 expression, remodeling of extracellular matrix, and increased directional migration of carrier cells to areas of pathology. This is an important finding since a consistent activation of MMP14 would enhance the directional migration of NSC carriers towards infiltrative tumor foci and would also increase the overall number of carriers that effectively reach and distribute within the malignant area. Such funding provides us the rational of modifying the NSC-based cell carrier to overexpress the molecule like MMP14 to enhance their tumor homing capacity.