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
  • enkephalin manufacturer In our current study IGFBP levels we

    2018-11-08

    In our current study IGFBP-2 levels were approximately 10-fold higher than those of IGFBP-3 suggesting that the large induction of IGFBP-2 may play a more import role in the differentiation of DPCs than the decrease seen in IGFBP-3 concentrations. Although an extensive literature describes both IGF-dependent and IGF independent effects of IGFBPs – reviewed in Annunziata et al. (2011) - in most instances quantitative aspects related to IGF and IGFBP concentrations in the local tissue environment have largely been ignored. Our data suggest that the ratio of IGF:IGFBP present in an experimental model can be important in determining the overall biological response (Fig. 4). These factors are also important with respect to the functional redundancy displayed by IGFBPs in some tissue culture and whole animal studies (Murphy, 1998; Pintar et al., 1995) and the accurate quantification of local IGF axis proteins in vivo or in vitro is essential for an understanding of biological outcomes. Notwithstanding these arguments evidence suggests an important role for the IGF axis in the differentiation and development of various dental structures. However there are several gaps in our knowledge of the role of various members of this molecular axis (particularly IGFBPs) in this process and it is hoped that future studies will shed some light in this area and also assist in design of strategies aimed at hard tissue engineering using multipotent stem enkephalin manufacturer derived from dental pulp and other stem cell niches within the oral cavity.
    Acknowledgements King AbdulAziz University – Jeddah (KAAU) HA, the King Faisal Specialist Hospital & research Centre – Jeddah (KFSH&RC-Jed) YH. RE acknowledges WELMEC, a Centre of Excellence in Medical Engineering funded by the Wellcome Trust and EPSRC, under grant number WT 088908/Z/09/Z for financial support. HA and YH acknowledge the Royal Embassy of Saudi Arabia – Cultural Bureau (UK) for financial support.
    Introduction In the adult SVZ, slow dividing type B stem cells give rise to rapidly dividing type C neural progenitor cells (or transit amplifying cells) that eventually differentiate into slow dividing type A neuroblasts (Doetsch et al., 1997, 1999; Aguirre et al., 2004). Neuroblasts are capable of migrating long distances, through a neurogenic region called the rostral migratory stream (RMS), to eventually terminally enkephalin manufacturer differentiate into interneurons in the olfactory bulb (Belluzzi et al., 2003). The RMS is a specialized migratory route that is ensheathed by astrocytes to form a tube-like structure, and is thought to provide instructive signals to the migrating neuroblasts (Sun et al., 2010). Migrating neuroblasts move in chains, where gap and adherens junctions between neighboring neuroblasts form connected movements (Lois et al., 1996; Ohab et al., 2006; Yamashita et al., 2006; Belvindrah et al., 2007; Kojima et al., 2010). After CNS injury and disease, neuroblasts have been shown to exit the RMS and migrate ectopically to sites of tissue damage (Arvidsson et al., 2002; Parent et al., 2002; Dixon et al., 2015); however, little is known about the cues that regulate ectopic migration. Outside the neurogenic regions neuroblasts have the capacity to differentiate into both neural and glial cell fates (Miragall et al., 1990; Bonfanti and Theodosis, 1994; Doetsch et al., 1997). In models of traumatic brain injury (TBI), migrating neuroblasts have been shown to provide beneficial effects to damaged tissues, such as the cortex, where they express trophic factors that support residential cell survival (Li et al., 2010; Dixon et al., 2015). In the absence of these cells, increased tissue damage and reduced functional recovery is observed (Dixon et al., 2015). Thus, expanding this population of cells and increasing their numbers in injured tissues can benefit functional recovery. Recent studies have shown that a family of receptor tyrosine kinase receptors, Eph receptors, and their ephrin ligands can regulate neural stem/progenitor cell (NSPC) and neuroblast proliferation and survival (Ricard et al., 2006; Furne et al., 2009; Theus et al., 2010; Theus et al., 2014). The ligand ephrinB3 is expressed in tissues surrounding the neurogenic niche, including by astrocytes that ensheathe the RMS (Ricard et al., 2006; Zhuang et al., 2010), whereas NSPCs express the EphB3 and EphA4 receptors but not ephrinB3 (Furne et al., 2009; Theus et al., 2010). Ephrins and Eph receptors are membrane-bound proteins that require direct cell-cell interaction to induce receptor signaling (Gale et al., 1996). B-class ephrins represent a subclass of ephrins that contain an intracellular domain also capable of signaling, such that interactions with their respective Eph receptor(s) can lead to bidirectional signals. Ephrins and Eph receptors are historically known to facilitate developmental tissue patterning and axon pathfinding through modulating actin cytoskeleton dynamics (Boyd et al., 2014). Activation of EphB3 and EphA4 by ephrinB3 can also induce anti-proliferative signs to negatively regulate the neurogenic response in both naïve and TBI conditions (Ricard et al., 2006; Furne et al., 2009; Theus et al., 2010; Theus et al., 2014). Here, we examine whether ephrinB3 regulates neuroblast migration in the TBI brain using loss-of-function and gain-of-function approaches, and demonstrate that ephrinB3 functions to restrict neuroblast movement to damaged tissues by limiting chain migration following injury.