Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Protoc ; 14(2): 331-349, 2019 02.
Article in English | MEDLINE | ID: mdl-30610242

ABSTRACT

Despite decades of research, pharmacological therapies for spinal cord motor pathologies are limited. Alternatives using macromolecular, viral, or cell-based therapies show early promise. However, introducing these substances into the spinal cord, past the blood-brain barrier, without causing injury is challenging. We describe a technique for intraspinal injection targeting the lumbar ventral horn in rodents. This technique preserves motor performance and has a proven track record of translation into phase 1 and 2 clinical trials in amyotrophic lateral sclerosis (ALS) patients. The procedure, in brief, involves exposure of the thoracolumbar spine and dissection of paraspinous muscles over the target vertebrae. Following laminectomy, the spine is affixed to a stereotactic frame, permitting precise and reproducible injection throughout the lumbar spine. We have used this protocol to inject various stem cell types, primarily human spinal stem cells (HSSCs); however, the injection is adaptable to any candidate therapeutic cell, virus, or macromolecule product. In addition to a detailed procedure, we provide stereotactic coordinates that assist in targeting of the lumbar spine and instructional videos. The protocol takes ~2 h per animal.


Subject(s)
Amyotrophic Lateral Sclerosis/surgery , Dissection/methods , Injections, Spinal/methods , Spinal Cord/surgery , Stereotaxic Techniques , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/therapy , Animals , Disease Models, Animal , Humans , Lumbosacral Region/surgery , Male , Mice, Transgenic , Motor Activity/physiology , Paraspinal Muscles/surgery , Rotarod Performance Test , Spinal Cord/pathology , Stem Cell Transplantation/methods , Transplantation, Heterologous
2.
Sci Rep ; 8(1): 14776, 2018 10 03.
Article in English | MEDLINE | ID: mdl-30283042

ABSTRACT

Stem cell transplantation offers a potentially transformative approach to treating neurodegenerative disorders. The safety of cellular therapies is established in multiple clinical trials, including our own in amyotrophic lateral sclerosis. To initiate similar trials in Alzheimer's disease, efficacious cell lines must be identified. Here, we completed a preclinical proof-of-concept study in the APP/PS1 murine model of Alzheimer's disease. Human neural stem cell transplantation targeted to the fimbria fornix significantly improved cognition in two hippocampal-dependent memory tasks at 4 and 16 weeks post-transplantation. While levels of synapse-related proteins and cholinergic neurons were unaffected, amyloid plaque load was significantly reduced in stem cell transplanted mice and associated with increased recruitment of activated microglia. In vitro, these same neural stem cells induced microglial activation and amyloid phagocytosis, suggesting an immunomodulatory capacity. Although long-term transplantation resulted in significant functional and pathological improvements in APP/PS1 mice, stem cells were not identified by immunohistochemistry or PCR at the study endpoint. These data suggest integration into native tissue or the idea that transient engraftment may be adequate for therapeutic efficacy, reducing the need for continued immunosuppression. Overall, our results support further preclinical development of human neural stem cells as a safe and effective therapy for Alzheimer's disease.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Peptides/genetics , Neural Stem Cells/pathology , Stem Cell Transplantation , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Cholinergic Neurons/metabolism , Cholinergic Neurons/pathology , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Humans , Immunosuppression Therapy/methods , Memory/physiology , Mice , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Phagocytosis/genetics , Synapses/genetics , Synapses/metabolism
3.
Ann Clin Transl Neurol ; 4(10): 749-755, 2017 10.
Article in English | MEDLINE | ID: mdl-29046883

ABSTRACT

The hippocampus has been the target of stem cell transplantations in preclinical studies focused on Alzheimer's disease, with results showing improvements in histological and behavioral outcomes. The corpus callosum is another structure that is affected early in Alzheimer's disease. Therefore, we hypothesize that this structure is a novel target for human neural stem cell transplantation in transgenic Alzheimer's disease mouse models. This study demonstrates the feasibility of targeting the corpus callosum and identifies an effective immunosuppression regimen for transplanted neural stem cell survival. These results support further preclinical development of the corpus callosum as a therapeutic target in Alzheimer's disease.

4.
Stem Cells Transl Med ; 5(3): 379-91, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26744412

ABSTRACT

Alzheimer's disease (AD) is the most prevalent age-related neurodegenerative disorder and a leading cause of dementia. Current treatment fails to modify underlying disease pathologies and very little progress has been made to develop effective drug treatments. Cellular therapies impact disease by multiple mechanisms, providing increased efficacy compared with traditional single-target approaches. In amyotrophic lateral sclerosis, we have shown that transplanted spinal neural stem cells (NSCs) integrate into the spinal cord, form synapses with the host, improve inflammation, and reduce disease-associated pathologies. Our current goal is to develop a similar "best in class" cellular therapy for AD. Here, we characterize a novel human cortex-derived NSC line modified to express insulin-like growth factor-I (IGF-I), HK532-IGF-I. Because IGF-I promotes neurogenesis and synaptogenesis in vivo, this enhanced NSC line offers additional environmental enrichment, enhanced neuroprotection, and a multifaceted approach to treating complex AD pathologies. We show that autocrine IGF-I production does not impact the cell secretome or normal cellular functions, including proliferation, migration, or maintenance of progenitor status. However, HK532-IGF-I cells preferentially differentiate into gamma-aminobutyric acid-ergic neurons, a subtype dysregulated in AD; produce increased vascular endothelial growth factor levels; and display an increased neuroprotective capacity in vitro. We also demonstrate that HK532-IGF-I cells survive peri-hippocampal transplantation in a murine AD model and exhibit long-term persistence in targeted brain areas. In conclusion, we believe that harnessing the benefits of cellular and IGF-I therapies together will provide the optimal therapeutic benefit to patients, and our findings support further preclinical development of HK532-IGF-I cells into a disease-modifying intervention for AD.


Subject(s)
Alzheimer Disease/therapy , Insulin-Like Growth Factor I/biosynthesis , Neural Stem Cells/transplantation , Neurogenesis , Alzheimer Disease/pathology , Animals , Cell Differentiation/genetics , Cell- and Tissue-Based Therapy , Disease Models, Animal , Gene Expression Regulation, Developmental , Humans , Insulin-Like Growth Factor I/genetics , Mice , Neural Stem Cells/cytology , Neurons/pathology , Neurons/transplantation , Synapses/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...