Your browser doesn't support javascript.
loading
Intranasally Administered L-Myc-Immortalized Human Neural Stem Cells Migrate to Primary and Distal Sites of Damage after Cortical Impact and Enhance Spatial Learning.
Gutova, Margarita; Cheng, Jeffrey P; Adhikarla, Vikram; Tsaturyan, Lusine; Barish, Michael E; Rockne, Russell C; Moschonas, Eleni H; Bondi, Corina O; Kline, Anthony E.
Afiliação
  • Gutova M; Department of Developmental & Stem Cell Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA.
  • Cheng JP; Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA.
  • Adhikarla V; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA.
  • Tsaturyan L; Department of Computational and Quantitative Medicine, Division of Mathematical Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA.
  • Barish ME; Department of Developmental & Stem Cell Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA.
  • Rockne RC; Department of Developmental & Stem Cell Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA.
  • Moschonas EH; Department of Computational and Quantitative Medicine, Division of Mathematical Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA.
  • Bondi CO; Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA.
  • Kline AE; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA.
Stem Cells Int ; 2021: 5549381, 2021.
Article em En | MEDLINE | ID: mdl-34122556
ABSTRACT
As the success of stem cell-based therapies is contingent on efficient cell delivery to damaged areas, neural stem cells (NSCs) have promising therapeutic potential because they inherently migrate to sites of central nervous system (CNS) damage. To explore the possibility of NSC-based therapy after traumatic brain injury (TBI), isoflurane-anesthetized adult male rats received a controlled cortical impact (CCI) of moderate severity (2.8 mm deformation at 4 m/s) or sham injury (i.e., no cortical impact). Beginning 1-week post-injury, the rats were immunosuppressed and 1 × 106 human NSCs (LM-NS008.GFP.fLuc) or vehicle (VEH) (2% human serum albumen) were administered intranasally (IN) on post-operative days 7, 9, 11, 13, 15, and 17. To evaluate the spatial distributions of the LM-NSC008 cells, half of the rats were euthanized on day 25, one day after completion of the cognitive task, and the other half were euthanized on day 46. 1 mm thick brain sections were optically cleared (CLARITY), and volumes were imaged by confocal microscopy. In addition, LM-NSC008 cell migration to the TBI site by immunohistochemistry for human-specific Nestin was observed at day 39. Acquisition of spatial learning was assessed in a well-established Morris water maze task on six successive days beginning on post-injury day 18. IN administration of LM-NSC008 cells after TBI (TBI + NSC) significantly facilitated spatial learning relative to TBI + VEH rats (p < 0.05) and had no effect on sham + NSC rats. Overall, these data indicate that IN-administered LM-NSC008 cells migrate to sites of TBI damage and that their presence correlates with cognitive improvement. Future studies will expand on these preliminary findings by evaluating other LM-NSC008 cell dosing paradigms and evaluating mechanisms by which LM-NSC008 cells contribute to cognitive recovery.

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2021 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2021 Tipo de documento: Article