induced neural stem cells
Recently Published Documents


TOTAL DOCUMENTS

70
(FIVE YEARS 28)

H-INDEX

15
(FIVE YEARS 2)

2022 ◽  
Vol 8 ◽  
Author(s):  
Warunya Chakritbudsabong ◽  
Ladawan Sariya ◽  
Phakhin Jantahiran ◽  
Nattarun Chaisilp ◽  
Somjit Chaiwattanarungruengpaisan ◽  
...  

The reprogramming of cells into induced neural stem cells (iNSCs), which are faster and safer to generate than induced pluripotent stem cells, holds tremendous promise for fundamental and frontier research, as well as personalized cell-based therapies for neurological diseases. However, reprogramming cells with viral vectors increases the risk of tumor development due to vector and transgene integration in the host cell genome. To circumvent this issue, the Sendai virus (SeV) provides an alternative integration-free reprogramming method that removes the danger of genetic alterations and enhances the prospects of iNSCs from bench to bedside. Since pigs are among the most successful large animal models in biomedical research, porcine iNSCs (piNSCs) may serve as a disease model for both veterinary and human medicine. Here, we report the successful generation of piNSC lines from pig fibroblasts by employing the SeV. These piNSCs can be expanded for up to 40 passages in a monolayer culture and produce neurospheres in a suspension culture. These piNSCs express high levels of NSC markers (PAX6, SOX2, NESTIN, and VIMENTIN) and proliferation markers (KI67) using quantitative immunostaining and western blot analysis. Furthermore, piNSCs are multipotent, as they are capable of producing neurons and glia, as demonstrated by their expressions of TUJ1, MAP2, TH, MBP, and GFAP proteins. During the reprogramming of piNSCs with the SeV, no induced pluripotent stem cells developed, and the established piNSCs did not express OCT4, NANOG, and SSEA1. Hence, the use of the SeV can reprogram porcine somatic cells without first going through an intermediate pluripotent state. Our research produced piNSCs using SeV methods in novel, easily accessible large animal cell culture models for evaluating the efficacy of iNSC-based clinical translation in human medicine. Additionally, our piNSCs are potentially applicable in disease modeling in pigs and regenerative therapies in veterinary medicine.


2022 ◽  
Vol 8 (1) ◽  
Author(s):  
Seung-Eun Lee ◽  
Daekee Kwon ◽  
Nari Shin ◽  
Dasom Kong ◽  
Nam Gyo Kim ◽  
...  

AbstractMitochondrial dysfunction is associated with familial Alzheimer’s disease (fAD), and the accumulation of damaged mitochondria has been reported as an initial symptom that further contributes to disease progression. In the amyloidogenic pathway, the amyloid precursor protein (APP) is cleaved by β-secretase to generate a C-terminal fragment, which is then cleaved by γ-secretase to produce amyloid-beta (Aβ). The accumulation of Aβ and its detrimental effect on mitochondrial function are well known, yet the amyloid precursor protein-derived C-terminal fragments (APP-CTFs) contributing to this pathology have rarely been reported. We demonstrated the effects of APP-CTFs-related pathology using induced neural stem cells (iNSCs) from AD patient-derived fibroblasts. APP-CTFs accumulation was demonstrated to mainly occur within mitochondrial domains and to be both a cause and a consequence of mitochondrial dysfunction. APP-CTFs accumulation also resulted in mitophagy failure, as validated by increased LC3-II and p62 and inconsistent PTEN-induced kinase 1 (PINK1)/E3 ubiquitin ligase (Parkin) recruitment to mitochondria and failed fusion of mitochondria and lysosomes. The accumulation of APP-CTFs and the causality of impaired mitophagy function were also verified in AD patient-iNSCs. Furthermore, we confirmed this pathological loop in presenilin knockout iNSCs (PSEN KO-iNSCs) because APP-CTFs accumulation is due to γ-secretase blockage and similarly occurs in presenilin-deficient cells. In the present work, we report that the contribution of APP-CTFs accumulation is associated with mitochondrial dysfunction and mitophagy failure in AD patient-iNSCs as well as PSEN KO-iNSCs.


2021 ◽  
Author(s):  
Warunya Chakritbudsabong ◽  
Ruttachuk Rungsiwiwut ◽  
Ladawan Sariya ◽  
Phakhin Juntahirun ◽  
Nattarun Chaisilp ◽  
...  

Abstract Background The reprogramming of cells to induced neural stem cells (iNSCs), faster and safer to generate than induced pluripotent stem cells, holds tremendous promise for disease modeling and personalized cell-based therapies for neurological diseases. Porcine iNSCs (piNSCs) may serve as a disease model for human medicine, as pigs are one of the most successful large animal models in biomedical research. Thus, this study aimed to establish safe and efficient integration-free piNSC lines.Methods The integration-free piNSC lines were generated by reprogramming porcine fibroblasts using the Sendai virus (SeV).Results Here we report the successful generation of integration-free piNSC lines using the SeV, with a reprogramming efficiency of 0.4%. The piNSCs can be expanded for up to 40 passages and express high levels of NSC markers (PAX6, NESTIN, and SOX2). They can produce neurons and glia, expressing TUJ, MAP2, TH, and GFAP. No induced pluripotent stem cells developed during reprogramming, and the established piNSCs did not express OCT4. Hence, the SeV can reprogram porcine fibroblast without first going through an intermediate pluripotent stage.Conclusions With the SeV approach, we generated integration-free piNSCs that may be used to assess the efficacy and safety of iNSC-based clinical translation in humans.


2021 ◽  
Vol 22 ◽  
Author(s):  
Saetbyul Hong ◽  
Seung-Eun Lee ◽  
Insung Kang ◽  
Jehoon Yang ◽  
Hunnyun Kim ◽  
...  

2021 ◽  
Vol 0 (0) ◽  
pp. 0-0
Author(s):  
Yuyan Liang ◽  
Sijie Li ◽  
Yujia Li ◽  
Mo Li ◽  
Xiaohong Sun ◽  
...  

2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii88-ii88
Author(s):  
Alison Mercer-Smith ◽  
Wulin Jiang ◽  
Alain Valdivia ◽  
Juli Bago ◽  
Scott Floyd ◽  
...  

Abstract INTRODUCTION Non-small cell lung cancer (NSCLC) is the most common cancer to form brain metastases. Radiation treatment is standard-of-care, but recurrence is still observed in 40% of patients. An adjuvant treatment is desperately needed to track down and kill tumor remnants after radiation. Tumoritropic neural stem cells (NSCs) that can home to and deliver a cytotoxic payload offer potential as such an adjuvant treatment. Here we show the transdifferentiation of human fibroblasts into tumor-homing induced neural stem cells (hiNSCs) that secrete the cytotoxic protein TRAIL (hiNSC-TRAIL) and explore the use of hiNSC-TRAIL to treat NSCLC brain metastases. METHODS To determine the migratory capacity of hiNSCs, hiNSCs were infused intracerebroventricularly (ICV) into mice bearing established bilateral NSCLC H460 brain tumors. hiNSC accumulation at tumor foci was monitored using bioluminescent imaging and post-mortem fluorescent analysis. To determine synergistic effects of radiation with TRAIL on NSCLC, we performed in vitro co-culture assays and isobologram analysis. In vivo, efficacy was determined by tracking the progression and survival of mice bearing intracranial H460 treated with hiNSC-TRAIL alone or in combination with 2 Gy radiation. RESULTS/CONCLUSION Following ICV infusion, hiNSCs persisted in the brain for > 1 week and migrated from the ventricles to colocalize with bilateral tumor foci. In vitro, viability assays and isobologram analysis revealed the combination treatment of hiNSC-TRAIL and 2 Gy radiation induced synergistic killing (combination index=0.64). In vivo, hiNSC-TRAIL/radiation combination therapy reduced tumor volumes > 90% compared to control-treated animals while radiation-only and hiNSC-TRAIL-only treated mice showed 21% and 52% reduced volumes, respectively. Dual-treatment extended survival 40%, increasing survival from a median of 20 days in controls to 28 days in the treatment group. These results suggest hiNSC-TRAIL can improve radiation therapy for NSCLC brain metastases and could potentially improve outcomes for patients suffering from this aggressive form of cancer.


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii98-ii98
Author(s):  
Hunter Bomba ◽  
Kevin Sheets ◽  
Alain Valdivia ◽  
Simon Khagi ◽  
Laura Ruterbories ◽  
...  

Abstract BACKGROUND Glioblastoma patient survival statistics have remained unchanged for more than three decades. Despite tumor resection and chemoradiotherapy, recurrence is inevitable. Moreover, the invasive behavior of glioblastoma confounds treatment. To improve patient survival statistics, a targeted therapy that can home to distant tumor foci is desperately needed. Induced neural stem cells (iNSCs) armed with cytotoxic payloads have proven efficacious against human xenograft models of glioblastoma. To further propel iNSCs to human clinical trials, we investigated the safety, toxicity, and persistence of iNSCs in a canine model. METHODS Autologous iNSCs generated from the skin of four non-tumor-bearing, purpose-bred, male beagles were engineered to express TRAIL and thymidine kinase (TK). iNSCs were loaded with ferumoxytol to facilitate MRI-tracking. Canines were divided into two cohorts to denote iNSC administration route: scaffold encapsulation or intracerebroventricular (ICV). Two dose levels were investigated: 1′106 iNSCs/kg or 3′106 iNSCs/kg. The scaffold cohort received a single dose of iNSCs while the ICV cohort received three doses of iNSCs via a Rickham reservoir. To activate TK, canines were administered valganciclovir. Canine health was assessed via neurological exams, MRI, and serial blood, urine, and CSF analyses. RESULTS No acute injection reactions were observed. Three of four canines exhibited surgery-induced blindness. Urine and CSF analyses were unremarkable. Unexpectedly, blood analyses showed transient neutropenia. Hypodense signal was observed on all MRI sequences through endpoint. Post-mortem histopathology of the spleen, liver, and lung were unremarkable. As expected, brain tissues exhibited gliosis, fibrous thickening, and inflammation. Spinal cords exhibited acute hemorrhaging, attributed to perimortem CSF draws. Surprisingly, significant testicular degeneration was observed; this was confirmed to be caused by valganciclovir. In conclusion, iNSCs exhibit limited toxicity and warrant further exploration. FUTURE DIRECTIONS Prospective studies will investigate the efficacy of autologous iNSCs in a spontaneous canine glioma model in preparation for human clinical trials.


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii89-ii89
Author(s):  
Andrew Buckley ◽  
Shaye Hagler ◽  
Vivien Lettry ◽  
Juli Bago ◽  
Spencer Maingi ◽  
...  

Abstract Induced neural stem-cells (iNSCs) represent a new opportunity in the emerging field of cellular immunotherapy. Patient-derived iNSCs modified to produce anti-tumoral compounds could lead to less rejection and safer outcomes than an off-the-shelf therapy. In this study, we established fibroblast lines (PFs) from skin-biopsies of patients being treated for glioblastoma (GBM) and transdifferentiated those fibroblasts into iNSC lines that produce anti-tumor compounds. We designed a combination of genomic and functional testing to assess iNSC line efficacy. Functional testing revealed differences in rate of transdifferentiation, therapeutic agent production, and tumor-homing amongst cell lines all of which varied among patients. RNAseq profiles of individual cells lines revealed biomarker signatures that differed in tumor-homing-pathways. There was no observed neuronal differentiation in the iNSCs from the transcriptomic profiles, indicating stability after transdifferentiation amongst PFs. Anti-tumor activity of patient-derived iNSCs was measured in vivo by surgical-resection mouse models with invasive CD133+ GBM cells. Patient-derived iNSCs showed variable tumoricidal effectiveness; more highly effective iNSC cells lines reduced tumor burden and increased survival post-resection from 28 to 45 days, whereas less effective cells lines could increase post-resection survival with increased iNSC dosage. PF origination and transcriptomic profile accounted for the differences amongst the iNSC lines. Identification of differentially expressed genes could indicate the cellular pathways that are most important for guaranteeing high, anti-tumoral activity. Further, the cellular profile of the patient-derived fibroblast influences the resulting cellular profile of the iNSC; potentially, correlation of fibroblast gene expression profile could predict tumoricidal efficacy of derived iNSCs.


Sign in / Sign up

Export Citation Format

Share Document