Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Sci Rep ; 13(1): 18439, 2023 10 27.
Article in English | MEDLINE | ID: mdl-37891179

ABSTRACT

Mucopolysaccharidosis III (MPSIII, Sanfilippo syndrome) is a devastating lysosomal storage disease that primarily affects the central nervous system. MPSIIIA is caused by loss-of-function mutations in the gene coding for sulfamidase (N-sulfoglucosamine sulfohydrolase/SGSH) resulting in SGSH enzyme deficiency, a buildup of heparin sulfate and subsequent neurodegeneration. There is currently no cure or disease modifying treatment for MPSIIIA. A mouse model for MPSIIIA was characterized in 1999 and later backcrossed onto the C57BL/6 background. In the present study, a novel immune deficient MPSIIIA mouse model (MPSIIIA-TKO) was created by backcrossing the immune competent, C57BL/6 MPSIIIA mouse to an immune deficient mouse model lacking Rag2, CD47 and Il2rg genes. The resulting mouse model has undetectable SGSH activity, exhibits histological changes consistent with MPSIIIA and lacks T cells, B cells and NK cells. This new mouse model has the potential to be extremely useful in testing human cellular therapies in an animal model as it retains the MPSIIIA disease phenotype while tolerating xenotransplantation.


Subject(s)
Mucopolysaccharidosis III , Animals , Humans , Mice , Mucopolysaccharidosis III/genetics , Mucopolysaccharidosis III/pathology , Mice, Inbred C57BL , Hydrolases/genetics , Phenotype , Disease Models, Animal
2.
Ann Clin Psychiatry ; 32(3): 182-196, 2020 08.
Article in English | MEDLINE | ID: mdl-32722729

ABSTRACT

BACKGROUND: NSI-189 phosphate (NSI-189) is a novel neurogenic molecule with pleiotropic properties, including antidepressant, procognitive, synaptoplastic, and neurotrophic activities demonstrated in preclinical studies. Its antidepressant activity is monoamine-independent. NSI-189 was previously tested in patients with recurrent major depressive disorder in an inpatient setting. METHODS: This study involved 220 patients randomized to an NSI-189 40-mg dose, NSI-189 80-mg dose, or placebo daily for 12 weeks. The study utilized the sequential parallel comparison design, in which the drug effect was tested in 2 separate stages of 6 weeks each. Herein, post-hoc analyses of the data are presented. RESULTS: NSI-189's antidepressant effect increased when the participants' initial baseline depression severity was dichotomized along a Montgomery-Åsberg Depression Rating Scale (MADRS) score of 30. The NSI-189 80-mg dose showed significant benefit over placebo when utilizing the MADRS-6 (P = .046) in the subgroup of patients who were moderately depressed (MADRS < 30) but was not significant in patients who were severely depressed (MADRS ≥30). More pronounced procognitive effects were also observed in the moderate subgroup relative to the severe subgroup or the whole study group, in which 11/36 (31%), 5/36 (14%), or 7/36 (19%) of CogScreen variables significantly improved, respectively. CONCLUSIONS: These results suggest that NSI-189 is effective as a safe adjunctive therapy, with most compelling antidepressant and procognitive benefits noted in patients with moderate depression.


Subject(s)
Aminopyridines/therapeutic use , Depressive Disorder, Major/drug therapy , Neurogenesis , Piperazines/therapeutic use , Adult , Female , Humans , Male , Middle Aged , Severity of Illness Index
3.
Oper Neurosurg (Hagerstown) ; 18(5): 503-510, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31414136

ABSTRACT

BACKGROUND: In preclinical studies, the Intracerebral Microinjection Instrument (IMI) has demonstrated the ability to deliver therapeutics within the brain in 3-dimensional arrays from a single overlying penetration while incurring minimal localized trauma. OBJECTIVE: To evaluate the safety and performance of the IMI in its first use in humans to deliver stem cells in complex configurations within brain regions affected by ischemic injury. METHODS: As part of a phase 1 study, 3 chronically hemiparetic motor stroke patients received intracerebral grafts of the therapeutic stem cell line, NSI-566, using the IMI and its supporting surgical planning software. The patients were 37 to 54 yr old, had ischemic strokes more than 1 yr prior to transplantation, and received Fugl-Meyer motor scale scores of 17-48 at screening. During a single surgical procedure, patients received several neural grafts (42 ± 3) within the peri-infarct region targeted strategically to facilitate neural repair. RESULTS: The IMI enabled multiple cellular deposits to be safely placed peripheral to stroke lesions. The procedure was well tolerated, recovery was uneventful, and there occurred no subsequent complications. The IMI performed reliably throughout the procedures without evident targeting errors. One year after transplantation, all 3 subjects displayed significant clinical improvement, and imaging analysis demonstrated occupation of infarct cavities with new tissue without tumor formation. CONCLUSION: IMI technology permits unprecedented numbers of injections to be tactically placed in 3-dimensional arrays safely and reliably in human subjects.This advanced methodology can optimize the benefits of novel therapeutics by enabling versatile 3-dimensional intracerebral targeting.


Subject(s)
Stroke , Brain , Humans , Microinjections , Stem Cell Transplantation , Stroke/diagnostic imaging , Stroke/surgery
4.
Stem Cells Transl Med ; 9(2): 177-188, 2020 02.
Article in English | MEDLINE | ID: mdl-31800978

ABSTRACT

Neural precursor cells (NSCs) hold great potential to treat a variety of neurodegenerative diseases and injuries to the spinal cord. However, current delivery techniques require an invasive approach in which an injection needle is advanced into the spinal parenchyma to deliver cells of interest. As such, this approach is associated with an inherent risk of spinal injury, as well as a limited delivery of cells into multiple spinal segments. Here, we characterize the use of a novel cell delivery technique that employs single bolus cell injections into the spinal subpial space. In immunodeficient rats, two subpial injections of human NSCs were performed in the cervical and lumbar spinal cord, respectively. The survival, distribution, and phenotype of transplanted cells were assessed 6-8 months after injection. Immunofluorescence staining and mRNA sequencing analysis demonstrated a near-complete occupation of the spinal cord by injected cells, in which transplanted human NSCs (hNSCs) preferentially acquired glial phenotypes, expressing oligodendrocyte (Olig2, APC) or astrocyte (GFAP) markers. In the outermost layer of the spinal cord, injected hNSCs differentiated into glia limitans-forming astrocytes and expressed human-specific superoxide dismutase and laminin. All animals showed normal neurological function for the duration of the analysis. These data show that the subpial cell delivery technique is highly effective in populating the entire spinal cord with injected NSCs, and has a potential for clinical use in cell replacement therapies for the treatment of ALS, multiple sclerosis, or spinal cord injury.


Subject(s)
Neural Stem Cells/metabolism , Parenchymal Tissue/metabolism , Animals , Parenchymal Tissue/cytology , Rats , Rats, Sprague-Dawley
5.
Diabetes ; 68(11): 2143-2154, 2019 11.
Article in English | MEDLINE | ID: mdl-31492662

ABSTRACT

While peripheral neuropathy is the most common complication of long-term diabetes, cognitive deficits associated with encephalopathy and myelopathy also occur. Diabetes is a risk factor for Alzheimer disease (AD) and increases the risk of progression from mild cognitive impairment to AD. The only current recommendation for preventing or slowing the progression of peripheral neuropathy is to maintain close glycemic control, while there is no recommendation for central nervous system disorders. NSI-189 is a new chemical entity that when orally administered promotes neurogenesis in the adult hippocampus, increases hippocampal volume, enhances synaptic plasticity, and reduces cognitive dysfunction. To establish the potential for impact on peripheral neuropathy, we first showed that NSI-189 enhances neurite outgrowth and mitochondrial functions in cultured adult rat primary sensory neurons. Oral delivery of NSI-189 to murine models of type 1 (female) and type 2 (male) diabetes prevented multiple functional and structural indices of small and large fiber peripheral neuropathy, increased hippocampal neurogenesis, synaptic markers and volume, and protected long-term memory. NSI-189 also halted progression of established peripheral and central neuropathy. NSI-189, which is currently in clinical trials for treatment of major depressive disorder, offers the opportunity for the development of a single therapeutic agent against multiple indices of central and peripheral neuropathy.


Subject(s)
Aminopyridines/therapeutic use , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Diabetic Neuropathies/drug therapy , Hippocampus/drug effects , Neurogenesis/drug effects , Piperazines/therapeutic use , Sensory Receptor Cells/drug effects , Aminopyridines/pharmacology , Animals , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 2/complications , Diabetic Neuropathies/physiopathology , Female , Male , Mice , Mitochondria/drug effects , Neuronal Outgrowth/drug effects , Piperazines/pharmacology , Rats , Synapses/drug effects
6.
Stem Cells Transl Med ; 8(10): 999-1007, 2019 10.
Article in English | MEDLINE | ID: mdl-31241246

ABSTRACT

NSI-566 is a stable, primary adherent neural stem cell line derived from a single human fetal spinal cord and expanded epigenetically with no genetic modification. This cell line is being tested in clinical trials in the U.S. for treatment of amyotrophic lateral sclerosis and spinal cord injury. In a single-site, phase I study, we evaluated the feasibility and safety of NSI-566 transplantation for the treatment of hemiparesis due to chronic motor stroke and determined the maximum tolerated dose for future trials. Three cohorts (n = 3 per cohort) were transplanted with one-time intracerebral injections of 1.2 × 107 , 2.4 × 107 , or 7.2 × 107 cells. Immunosuppression therapy with tacrolimus was maintained for 28 days. All subjects had sustained chronic motor strokes, verified by magnetic resonance imaging (MRI), initiated between 5 and 24 months prior to surgery with modified Rankin Scores [MRSs] of 2, 3, or 4 and Fugl-Meyer Motor Scores of 55 or less. At the 12-month visit, the mean Fugl-Meyer Motor Score (FMMS, total score of 100) for the nine participants showed 16 points of improvement (p = .0078), the mean MRS showed 0.8 points of improvement (p = .031), and the mean National Institutes of Health Stroke Scale showed 3.1 points of improvement (p = .020). For six participants who were followed up for 24 months, these mean changes remained stable. The treatment was well tolerated at all doses. Longitudinal MRI studies showed evidence indicating cavity-filling by new neural tissue formation in all nine patients. Although this was a small, one-arm study of feasibility, the results are encouraging to warrant further studies. Stem Cells Translational Medicine 2019;8:999-1007.


Subject(s)
Brain Ischemia/complications , Brain Ischemia/therapy , Neural Stem Cells/transplantation , Paralysis/therapy , Stroke/complications , Stroke/therapy , Adult , Aged , Female , Humans , Male , Middle Aged , Treatment Outcome
7.
Front Neurol ; 10: 82, 2019.
Article in English | MEDLINE | ID: mdl-30809187

ABSTRACT

Human neural stem cells (hNSCs) transplantation in several brain injury models has established their therapeutic potential. However, the feasibility of hNSCs transplantation is still not clear for acute subdural hematoma (ASDH) brain injury that needs external decompression. Thus, the aim of this pilot study was to test feasibility using a rat ASDH decompression model with two clinically relevant transplantation methods. Two different methods, in situ stereotactic injection and hNSC-embedded matrix seating on the brain surface, were attempted. Athymic rats were randomized to uninjured or ASDH groups (F344/NJcl-rnu/rnu, n = 7-10/group). Animals in injury group were subjected to ASDH, and received decompressive craniectomy and 1-week after decompression surgery were transplanted with green fluorescent protein (GFP)-transduced hNSCs using one of two approaches. Histopathological examinations at 4 and 8 weeks showed that the GFP-positive hNSCs survived in injured brain tissue, extended neurite-like projections resembling neural dendrites. The in situ transplantation group had greater engraftment of hNSCs than matrix embedding approach. Immunohistochemistry with doublecortin, NeuN, and GFAP at 8 weeks after transplantation showed that transplanted hNSCs remained as immature neurons and did not differentiate toward to glial cell lines. Motor function was assessed with rotarod, compared to control group (n = 10). The latency to fall from the rotarod in hNSC in situ transplanted rats was significantly higher than in control rats (median, 113 s in hNSC vs. 69 s in control, P = 0.02). This study first demonstrates the robust engraftment of in situ transplanted hNSCs in a clinically-relevant ASDH decompression rat model. Further preclinical studies with longer study duration are warranted to verify the effectiveness of hNSC transplantation in amelioration of TBI induced deficits.

8.
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
9.
Neuropharmacology ; 144: 337-344, 2019 01.
Article in English | MEDLINE | ID: mdl-30408487

ABSTRACT

NSI-189 Phosphate, (4-benzylpiperazin-1-yl)-[2-(3-methyl-butylamino)pyridin-3-yl] methanone is a new chemical entity under development for the treatment of MDD, based upon preclinical data demonstrating stimulation of neurogenesis of human hippocampus-derived neural stem cells in vitro and in mouse hippocampus in vivo. Previous studies have examined the tolerability and efficacy of NSI-189 for treating major depressive disorder (MDD). NSI-189 has shown significant potential as a treatment for MDD, with concurrent improvement of a cognition scale in a small double-blind, placebo-controlled study. The current study evaluated its possible application for the treatment of Angelman Syndrome. Incubation of acute hippocampal slices from wild-type mice with NSI-189 resulted in a time- and dose-dependent increase in the magnitude of long-term potentiation (LTP) elicited by theta burst stimulation (TBS). The same protocol enhanced TBS-induced LTP in acute hippocampal slices from AS mice. A short treatment with daily injections of NSI-189 in AS mice reversed impairments in cognitive and motor functions, while it slightly enhanced performance of WT mice. The effects of NSI-189 on synaptic plasticity and cognitive functions were associated with activation of the TrkB and Akt pathways. These results suggest that NSI-189 could represent a potential treatment for AS patients.


Subject(s)
Aminopyridines/pharmacology , Angelman Syndrome/drug therapy , Central Nervous System Agents/pharmacology , Cognition/drug effects , Hippocampus/drug effects , Long-Term Potentiation/drug effects , Piperazines/pharmacology , Angelman Syndrome/physiopathology , Angelman Syndrome/psychology , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Hippocampus/physiopathology , Male , Mice, Transgenic , Motor Activity/drug effects , Tissue Culture Techniques
10.
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
11.
Ann Clin Transl Neurol ; 5(6): 730-740, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29928656

ABSTRACT

OBJECTIVE: Intraspinal human spinal cord-derived neural stem cell (HSSC) transplantation is a potential therapy for amyotrophic lateral sclerosis (ALS); however, previous trials lack controls. This post hoc analysis compared ambulatory limb-onset ALS participants in Phase 1 and 2 (Ph1/2) open-label intraspinal HSSC transplantation studies up to 3 years after transplant to matched participants in Pooled Resource Open-Access ALS Clinical Trials (PRO-ACT) and ceftriaxone datasets to provide required analyses to inform future clinical trial designs. METHODS: Survival, ALSFRS-R, and a composite statistic (ALS/SURV) combining survival and ALS Functional Rating Scale revised (ALSFRS-R) functional status were assessed for matched participant subsets: PRO-ACT n = 1108, Ph1/2 n = 21 and ceftriaxone n = 177, Ph1/2 n = 20. RESULTS: Survival did not differ significantly between cohorts: Ph1/2 median survival 4.7 years, 95% CI (1.2, ∞) versus PRO-ACT 2.3 years (1.9, 2.5), P = 1.0; Ph1/2 3.0 years (1.2, 5.6) versus ceftriaxone 2.3 years (1.8, 2.8), P = 0.88. Mean ALSFRS-R at 24 months significantly differed between Ph1/2 and both comparison cohorts (Ph1/2 30.1 ± 8.6 vs. PRO-ACT 24.0 ± 10.2, P = 0.048; Ph1/2 30.7 ± 8.8 vs. ceftriaxone 19.2 ± 9.5, P = 0.0023). Using ALS/SURV, median PRO-ACT and ceftriaxone participants died by 24 months, whereas median Ph1/2 participant ALSFRS-Rs were 23 (P = 0.0038) and 19 (P = 0.14) in PRO-ACT and ceftriaxone comparisons at 24 months, respectively, supporting improved functional outcomes in the Ph1/2 study. INTERPRETATION: Comparison of Ph1/2 studies to historical datasets revealed significantly improved survival and function using ALS/SURV versus PRO-ACT controls. While results are encouraging, comparison against historical populations demonstrate limitations in noncontrolled studies. These findings support continued evaluation of HSSC transplantation in ALS, support the benefit of control populations, and enable necessary power calculations to design a randomized, sham surgery-controlled efficacy study.

12.
Radiat Res ; 189(4): 345-353, 2018 04.
Article in English | MEDLINE | ID: mdl-29351056

ABSTRACT

Clinical management of primary and secondary central nervous system (CNS) malignancies frequently includes radiotherapy to forestall tumor growth and recurrence after surgical resection. While cranial radiotherapy remains beneficial, adult and pediatric brain tumor survivors suffer from a wide range of debilitating and progressive cognitive deficits. Although this has been recognized as a significant problem for decades, there remains no clinical recourse for the unintended neurocognitive sequelae associated with these types of cancer treatments. In previous work, multiple mechanisms have been identified that contribute to radiation-induced cognitive dysfunction, including the inhibition of neurogenesis caused by the depletion of radiosensitive populations of stem and progenitor cells in the hippocampus. To explore the potential neuroprotective properties of a pro-neurogenic compound NSI-189, Long-Evans rats were subjected to a clinically relevant fractionated irradiation protocol followed by four weeks of NSI-189 administered daily by oral gavage. Animals were then subjected to five different behavioral tasks followed by an analysis of neurogenesis, hippocampal volume and neuroinflammation. Irradiated cohorts manifested significant behavioral decrements on all four spontaneous exploration tasks. Importantly, NSI-189 treatment resulted in significantly improved performance in four of these tasks: novel place recognition, novel object recognition, object in place and temporal order. In addition, there was a trend of improved performance in the contextual phase of the fear conditioning task. Importantly, enhanced cognition in the NSI-189-treated cohort was found to persist one month after the cessation of drug treatment. These neurocognitive benefits of NSI-189 coincided with a significant increase in neurogenesis and a significant decrease in the numbers of activated microglia compared to the irradiated cohort that was given vehicle alone. The foregoing changes were not accompanied by major changes in hippocampal volume. These data demonstrate that oral administration of a pro-neurogenic compound exhibiting anti-inflammatory indications could impart long-term neurocognitive benefits in the irradiated brain.


Subject(s)
Aminopyridines/administration & dosage , Aminopyridines/pharmacology , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Piperazines/administration & dosage , Piperazines/pharmacology , Radiation Injuries, Experimental/drug therapy , Radiation Injuries, Experimental/etiology , Administration, Oral , Animals , Cognition/drug effects , Cognition/radiation effects , Cognitive Dysfunction/pathology , Cognitive Dysfunction/physiopathology , Conditioning, Psychological/drug effects , Conditioning, Psychological/radiation effects , Cranial Irradiation/adverse effects , Fear/psychology , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Hippocampus/radiation effects , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Organ Size/drug effects , Organ Size/radiation effects , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/physiopathology , Rats , Recognition, Psychology/drug effects , Recognition, Psychology/radiation effects
13.
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.

14.
Expert Opin Investig Drugs ; 26(6): 767-770, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28460574

ABSTRACT

INTRODUCTION: Alterations in neurogenic and neurotrophic processes as well as intracellular signalling cascades provides the basis for hypothesizing that neurogenic agents may be therapeutic across multiple RDoC-defined domains (e.g. positive valence systems, general cognitive processes). Moreover, using the DSM-5 taxonomy, neurogenic agents may mitigate symptoms in adults with depressive and bipolar disorders as well as individuals with cognitive disorders. Areas covered: NSI-189 is a benzylpiperizine-aminiopyridine, a novel chemical entity that stimulates neurogenesis of human hippocampus-derived neural stem cells in vitro and stimulates neurogenesis in murine hippocampus in vivo. Emerging evidence also indicates that NSI-189 phosphate has regionally specific effects insofar as neurogenesis is observed largely in the subventricular zone of the hippocampus. Results from a preliminary proof of concept study suggests that NSI-189 may be capable of mitigating depressive symptoms and improve cognitive function in adults with DSM-5-defined Major Depressive Disorder (MDD). Expert opinion: Preliminary proof-of-concept studies indicate both antidepressant and procognitive effects. Beneficial effects in cognitive-emotional processing as well as whether the procognitive effects are independent of antidepressant effects are vistas of future research. Taken together, NSI-189 is a multi-domain neurogenic compound with brain-therapeutic properties with potential therapeutic applications across disparate psychiatric disorders.


Subject(s)
Aminopyridines/pharmacology , Cognition Disorders/drug therapy , Depressive Disorder, Major/drug therapy , Piperazines/pharmacology , Adult , Animals , Antidepressive Agents/pharmacology , Bipolar Disorder/drug therapy , Bipolar Disorder/physiopathology , Cognition/drug effects , Cognition Disorders/physiopathology , Depressive Disorder, Major/physiopathology , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Neurogenesis/drug effects
15.
J Neurotrauma ; 34(11): 1981-1995, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28249550

ABSTRACT

Penetrating traumatic brain injury (PTBI) is one of the major cause of death and disability worldwide. Previous studies with penetrating ballistic-like brain injury (PBBI), a PTBI rat model revealed widespread perilesional neurodegeneration, similar to that seen in humans following gunshot wound to the head, which is unmitigated by any available therapies to date. Therefore, we evaluated human neural stem cell (hNSC) engraftment to putatively exploit the potential of cell therapy that has been seen in other central nervous system injury models. Toward this objective, green fluorescent protein (GFP) labeled hNSC (400,000 per animal) were transplanted in immunosuppressed Sprague-Dawley (SD), Fisher, and athymic (ATN) PBBI rats 1 week after injury. Tacrolimus (3 mg/kg 2 days prior to transplantation, then 1 mg/kg/day), methylprednisolone (10 mg/kg on the day of transplant, 1 mg/kg/week thereafter), and mycophenolate mofetil (30 mg/kg/day) for 7 days following transplantation were used to confer immunosuppression. Engraftment in SD and ATN was comparable at 8 weeks post-transplantation. Evaluation of hNSC differentiation and distribution revealed increased neuronal differentiation of transplanted cells with time. At 16 weeks post-transplantation, neither cell proliferation nor glial lineage markers were detected. Transplanted cell morphology was similar to that of neighboring host neurons, and there was relatively little migration of cells from the peritransplant site. By 16 weeks, GFP-positive processes extended both rostrocaudally and bilaterally into parenchyma, spreading along host white matter tracts, traversing the internal capsule, and extending ∼13 mm caudally from transplantation site reaching into the brainstem. In a Morris water maze test at 8 weeks post-transplantation, animals with transplants had shorter latency to platform than vehicle-treated animals. However, weak injury-induced cognitive deficits in the control group at the delayed time point confounded benefits of durable engraftment and neuronal differentiation. Therefore, these results justify further studies to progress towards clinical translation of hNSC therapy for PTBI.


Subject(s)
Cell Differentiation/physiology , Cognition Disorders/therapy , Head Injuries, Penetrating/therapy , Neural Stem Cells/transplantation , Neurons/physiology , Stem Cell Transplantation/methods , Animals , Brain Injuries, Traumatic/diagnosis , Brain Injuries, Traumatic/therapy , Cognition Disorders/diagnosis , Head Injuries, Penetrating/diagnosis , Humans , Random Allocation , Rats , Rats, Inbred F344 , Rats, Nude , Rats, Sprague-Dawley
16.
J Cell Physiol ; 232(10): 2731-2740, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28181668

ABSTRACT

Enhancing neurogenesis may be a powerful stroke therapy. Here, we tested in a rat model of ischemic stroke the beneficial effects of NSI-189, an orally active, new molecular entity (mol. wt. 366) with enhanced neurogenic activity, and indicated as an anti-depressant drug in a clinical trial (Fava et al., , Molecular Psychiatry, DOI: 10.1038/mp.2015.178) and being tested in a Phase 2 efficacy trial (ClinicalTrials.gov, , ClinicalTrials.gov Identifier: NCT02695472) for treatment of major depression. Oral administration of NSI-189 in adult Sprague-Dawley rats starting at 6 hr after middle cerebral artery occlusion, and daily thereafter over the next 12 weeks resulted in significant amelioration of stroke-induced motor and neurological deficits, which was maintained up to 24 weeks post-stroke. Histopathological assessment of stroke brains from NSI-189-treated animals revealed significant increments in neurite outgrowth as evidenced by MAP2 immunoreactivity that was prominently detected in the hippocampus and partially in the cortex. These results suggest NSI-189 actively stimulated remodeling of the stroke brain. Parallel in vitro studies further probed this remodeling process and demonstrated that oxygen glucose deprivation and reperfusion (OGD/R) initiated typical cell death processes, which were reversed by NSI-189 treatment characterized by significant attenuation of OGD/R-mediated hippocampal cell death and increased Ki67 and MAP2 expression, coupled with upregulation of neurogenic factors such as BDNF and SCF. These findings support the use of oral NSI-189 as a therapeutic agent well beyond the initial 6-hr time window to accelerate and enhance the overall functional improvement in the initial 6 months post stroke.


Subject(s)
Aminopyridines/pharmacology , Behavior, Animal/drug effects , Brain/drug effects , Infarction, Middle Cerebral Artery/drug therapy , Motor Activity/drug effects , Neurogenesis/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Piperazines/pharmacology , Animals , Brain/metabolism , Brain/pathology , Brain/physiopathology , Brain-Derived Neurotrophic Factor/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/psychology , Ki-67 Antigen/metabolism , Male , Microtubule-Associated Proteins/metabolism , Neuronal Outgrowth/drug effects , Neurons/metabolism , Neurons/pathology , Rats, Sprague-Dawley , Recovery of Function , Time Factors
17.
Neuromodulation ; 19(7): 708-716, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27593216

ABSTRACT

OBJECTIVES: The ability to safely place viable intracerebral grafts of human-derived therapeutic stem cells in three-dimensional (3D) space was assessed in a porcine model of human stereotactic surgery using the Intracerebral Microinjection Instrument (IMI) compared to a conventional straight cannula. MATERIALS AND METHODS: Two groups of healthy minipigs received injections of the human stem cell line, NSI-566, into the right hemisphere and cell suspension carrier media into the left hemisphere. Group A received all injections using a straight, 21-gauge stainless steel cannula. Group B received all injections using the IMI, whereby radial distribution of injections was achieved via angular extension of a 196-micron diameter cannula from a single overlying penetration of the guide cannula. Each animal received six 20 µL intracerebral-injections within each hemisphere: three in a radial distribution, covering a 180° arc with each injection separated by a 60° arc distance, within both frontal cortex and basal ganglia. H&E and immunocytochemistry (HuNu and GFAP) were used to identify implanted cells and to assess tissue response. RESULTS: The presence of surviving cells in appropriate brain regions demonstrated that the IMI is capable of accurately delivering viable human-derived stem cells safely in a 3D array at predetermined sites within the pig brain. In addition, qualitative evaluation of the target tissue suggests efficient delivery with decreased surgical trauma. CONCLUSIONS: In contrast to traditional straight cannulas, the IMI enables the delivery of multiple precise cellular injection volumes in accurate 3D arrays. In this porcine large animal model of human neurosurgery, the IMI reduced surgical time and appeared to reduce neural trauma associated with multiple penetrations that would otherwise be required using a conventional straight delivery cannula.


Subject(s)
Brain Injuries, Traumatic/surgery , Embryonic Stem Cells/physiology , Microinjections/methods , Stem Cell Transplantation/methods , Animals , Brain/diagnostic imaging , Brain/metabolism , Brain/pathology , Brain Injuries, Traumatic/diagnostic imaging , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Humans , Imaging, Three-Dimensional , Magnetic Resonance Imaging , Phosphopyruvate Hydratase/metabolism , Swine , Swine, Miniature
18.
Neurology ; 87(4): 392-400, 2016 Jul 26.
Article in English | MEDLINE | ID: mdl-27358335

ABSTRACT

OBJECTIVE: To test the safety of spinal cord transplantation of human stem cells in patients with amyotrophic lateral sclerosis (ALS) with escalating doses and expansion of the trial to multiple clinical centers. METHODS: This open-label trial included 15 participants at 3 academic centers divided into 5 treatment groups receiving increasing doses of stem cells by increasing numbers of cells/injection and increasing numbers of injections. All participants received bilateral injections into the cervical spinal cord (C3-C5). The final group received injections into both the lumbar (L2-L4) and cervical cord through 2 separate surgical procedures. Participants were assessed for adverse events and progression of disease, as measured by the ALS Functional Rating Scale-Revised, forced vital capacity, and quantitative measures of strength. Statistical analysis focused on the slopes of decline of these phase 2 trial participants alone or in combination with the phase 1 participants (previously reported), comparing these groups to 3 separate historical control groups. RESULTS: Adverse events were mostly related to transient pain associated with surgery and to side effects of immunosuppressant medications. There was one incident of acute postoperative deterioration in neurologic function and another incident of a central pain syndrome. We could not discern differences in surgical outcomes between surgeons. Comparisons of the slopes of decline with the 3 separate historical control groups showed no differences in mean rates of progression. CONCLUSIONS: Intraspinal transplantation of human spinal cord-derived neural stem cells can be safely accomplished at high doses, including successive lumbar and cervical procedures. The procedure can be expanded safely to multiple surgical centers. CLASSIFICATION OF EVIDENCE: This study provides Class IV evidence that for patients with ALS, spinal cord transplantation of human stem cells can be safely accomplished and does not accelerate the progression of the disease. This study lacks the precision to exclude important benefit or safety issues.


Subject(s)
Amyotrophic Lateral Sclerosis/therapy , Neural Stem Cells/transplantation , Spinal Cord/surgery , Stem Cell Transplantation/methods , Adult , Age of Onset , Cervical Vertebrae , Female , Humans , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/therapeutic use , Lumbosacral Region , Male , Middle Aged , Stem Cell Transplantation/adverse effects , Treatment Outcome
19.
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
20.
Stem Cells ; 33(5): 1480-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25532472

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder resulting in motor neuron (MN) loss. There are currently no effective therapies; however, cellular therapies using neural progenitor cells protect MNs and attenuate disease progression in G93A-SOD1 ALS rats. Recently, we completed a phase I clinical trial examining intraspinal human spinal stem cell (HSSC) transplantation in ALS patients which demonstrated our approach was safe and feasible, supporting the phase II trial currently in progress. In parallel, efforts focused on understanding the mechanisms underlying the preclinical benefit of HSSCs in vitro and in animal models of ALS led us to investigate how insulin-like growth factor-I (IGF-I) production contributes to cellular therapy neuroprotection. IGF-I is a potent growth factor with proven efficacy in preclinical ALS studies, and we contend that autocrine IGF-I production may enhance the salutary effects of HSSCs. By comparing the biological properties of HSSCs to HSSCs expressing sixfold higher levels of IGF-I, we demonstrate that IGF-I production augments the production of glial-derived neurotrophic factor and accelerates neurite outgrowth without adversely affecting HSSC proliferation or terminal differentiation. Furthermore, we demonstrate that increased IGF-I induces more potent MN protection from excitotoxicity via both indirect and direct mechanisms, as demonstrated using hanging inserts with primary MNs or by culturing with organotypic spinal cord slices, respectively. These findings support our theory that combining autocrine growth factor production with HSSC transplantation may offer a novel means to achieve additive neuroprotection in ALS.


Subject(s)
Autocrine Communication , Insulin-Like Growth Factor I/metabolism , Neural Stem Cells/metabolism , Neuroprotection , Animals , Cell Differentiation , Cell Movement , Cell Proliferation , Humans , Neuroprotective Agents/metabolism , Rats , Real-Time Polymerase Chain Reaction , Receptor, IGF Type 1/metabolism , Spinal Cord/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...