Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Sci Transl Med ; 10(440)2018 05 09.
Article in English | MEDLINE | ID: mdl-29743351

ABSTRACT

The use of autologous (or syngeneic) cells derived from induced pluripotent stem cells (iPSCs) holds great promise for future clinical use in a wide range of diseases and injuries. It is expected that cell replacement therapies using autologous cells would forego the need for immunosuppression, otherwise required in allogeneic transplantations. However, recent studies have shown the unexpected immune rejection of undifferentiated autologous mouse iPSCs after transplantation. Whether similar immunogenic properties are maintained in iPSC-derived lineage-committed cells (such as neural precursors) is relatively unknown. We demonstrate that syngeneic porcine iPSC-derived neural precursor cell (NPC) transplantation to the spinal cord in the absence of immunosuppression is associated with long-term survival and neuronal and glial differentiation. No tumor formation was noted. Similar cell engraftment and differentiation were shown in spinally injured transiently immunosuppressed swine leukocyte antigen (SLA)-mismatched allogeneic pigs. These data demonstrate that iPSC-NPCs can be grafted into syngeneic recipients in the absence of immunosuppression and that temporary immunosuppression is sufficient to induce long-term immune tolerance after NPC engraftment into spinally injured allogeneic recipients. Collectively, our results show that iPSC-NPCs represent an alternative source of transplantable NPCs for the treatment of a variety of disorders affecting the spinal cord, including trauma, ischemia, or amyotrophic lateral sclerosis.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/transplantation , Spinal Cord/transplantation , Aging , Animals , Cell Differentiation , Cellular Reprogramming , Chronic Disease , Fibroblasts/cytology , Gene Expression Regulation , Immune Tolerance , Immunity, Humoral , Immunosuppression Therapy , Neostriatum/pathology , Neural Stem Cells/cytology , Neurons/cytology , Rats , Skin/cytology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/therapy , Survival Analysis , Swine , Swine, Miniature , Transplantation, Homologous , Transplantation, Isogeneic
2.
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
3.
J Comp Neurol ; 522(12): 2784-801, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24610493

ABSTRACT

An important component for successful translation of cell replacement-based therapies into clinical practice is the utilization of large animal models to conduct efficacy and/or safety cell dosing studies. Over the past few decades, several large animal models (dog, cat, nonhuman primate) were developed and employed in cell replacement studies; however, none of these models appears to provide a readily available platform to conduct effective and large-scale preclinical studies. In recent years, numerous pig models of neurodegenerative disorders were developed using both a transgenic approach as well as invasive surgical techniques. The pig model (naïve noninjured animals) was recently used successfully to define the safety and optimal dosing of human spinal stem cells after grafting into the central nervous system (CNS) in immunosuppressed animals. The data from these studies were used in the design of a human clinical protocol used in amyotrophic lateral sclerosis (ALS) patients in a Phase I clinical trial. In addition, a highly inbred (complete major histocompatibility complex [MHC] match) strain of miniature pigs is available which permits the design of comparable MHC combinations between the donor cells and the graft recipient as used in human patients. Jointly, these studies show that the pig model can represent an effective large animal model to be used in preclinical cell replacement modeling. This review summarizes the available pig models of neurodegenerative disorders and the use of some of these models in cell replacement studies. The challenges and potential future directions in more effective use of the pig neurodegenerative models are also discussed.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Disease Models, Animal , Neurodegenerative Diseases/surgery , Animals , Humans , Swine
4.
Pain ; 155(6): 1150-1160, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24631588

ABSTRACT

PI3-kinases (PI3Ks) participate in nociception within spinal cord, dorsal root ganglion (DRG), and peripheral nerves. To extend our knowledge, we immunohistochemically stained for each of the 4 class I PI3K isoforms along with several cell-specific markers within the lumbar spinal cord, DRG, and sciatic nerve of naive rats. Intrathecal and intraplantar isoform specific antagonists were given as pretreatments before intraplantar carrageenan; pain behavior was then assessed over time. The α-isoform was localized to central terminals of primary afferent fibers in spinal cord laminae IIi to IV as well as to neurons in ventral horn and DRG. The PI3Kß isoform was the only class I isoform seen in dorsal horn neurons; it was also observed in DRG, Schwann cells, and axonal paranodes. The δ-isoform was found in spinal cord white matter oligodendrocytes and radial astrocytes, and the γ-isoform was seen in a subpopulation of IB4-positive DRG neurons. No isoform co-localized with microglial markers or satellite cells in naive tissue. Only the PI3Kß antagonist, but none of the other antagonists, had anti-allodynic effects when administered intrathecally; coincident with reduced pain behavior, this agent completely blocked paw carrageenan-induced dorsal horn 2-amino-3-(3-hydroxy-5-methyl-isoxazol-4-yl) propanoic acid (AMPA) receptor trafficking to plasma membranes. Intraplantar administration of the γ-antagonist prominently reduced pain behavior. These data suggest that each isoform displays specificity with regard to neuronal type as well as to specific tissues. Furthermore, each PI3K isoform has a unique role in development of nociception and tissue inflammation.


Subject(s)
Acute Pain/enzymology , Ganglia, Spinal/enzymology , Phosphatidylinositol 3-Kinase/physiology , Spinal Cord/enzymology , Acute Pain/pathology , Animals , Ganglia, Spinal/chemistry , Ganglia, Spinal/pathology , Inflammation/enzymology , Inflammation/pathology , Isoenzymes/analysis , Isoenzymes/physiology , Male , Phosphatidylinositol 3-Kinase/analysis , Rats , Rats, Sprague-Dawley , Spinal Cord/chemistry , Spinal Cord/pathology
5.
Exp Neurol ; 248: 85-99, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23748136

ABSTRACT

Achievement of effective, safe and long-term immunosuppression represents one of the challenges in experimental allogeneic and xenogeneic cell and organ transplantation. The goal of the present study was to develop a reliable, long-term immunosuppression protocol in Sprague-Dawley (SD) rats by: 1) comparing the pharmacokinetics of four different subcutaneously delivered/implanted tacrolimus (TAC) formulations, including: i) caster oil/saline solution, ii) unilamellar or multilamellar liposomes, iii) biodegradable microspheres, and iv) biodegradable 3-month lasting pellets; and 2) defining the survival and immune response in animals receiving spinal injections of human neural precursors at 6 weeks to 3 months after cell grafting. In animals implanted with TAC pellets (3.4 mg/kg/day), a stable 3-month lasting plasma concentration of TAC averaging 19.1 ± 4.9 ng/ml was measured. Analysis of grafted cell survival in SOD+ or spinal trauma-injured SD rats immunosuppressed with 3-month lasting TAC pellets (3.4-5.1 mg/kg/day) showed the consistent presence of implanted human neurons with minimal or no local T-cell infiltration. These data demonstrate that the use of TAC pellets can represent an effective, long-lasting immunosuppressive drug delivery system that is safe, simple to implement and is associated with a long-term human neural precursor survival after grafting into the spinal cord of SOD+ or spinal trauma-injured SD rats.


Subject(s)
Graft Survival/drug effects , Immunosuppression Therapy/methods , Immunosuppressive Agents/administration & dosage , Neural Stem Cells/transplantation , Spinal Cord/drug effects , Tacrolimus/administration & dosage , Animals , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Drug Implants , Graft Survival/immunology , Humans , Immunosuppressive Agents/pharmacokinetics , Neural Stem Cells/immunology , Neurons/immunology , Neurons/transplantation , Rats , Rats, Sprague-Dawley , Spinal Cord/immunology , Spinal Cord Injuries/immunology , Tacrolimus/pharmacokinetics
6.
PLoS One ; 7(8): e42614, 2012.
Article in English | MEDLINE | ID: mdl-22916141

ABSTRACT

BACKGROUND: Mutation in the ubiquitously expressed cytoplasmic superoxide dismutase (SOD1) causes an inherited form of Amyotrophic Lateral Sclerosis (ALS). Mutant synthesis in motor neurons drives disease onset and early disease progression. Previous experimental studies have shown that spinal grafting of human fetal spinal neural stem cells (hNSCs) into the lumbar spinal cord of SOD1(G93A) rats leads to a moderate therapeutical effect as evidenced by local α-motoneuron sparing and extension of lifespan. The aim of the present study was to analyze the degree of therapeutical effect of hNSCs once grafted into the lumbar spinal ventral horn in presymptomatic immunosuppressed SOD1(G93A) rats and to assess the presence and functional integrity of the descending motor system in symptomatic SOD1(G93A) animals. METHODS/PRINCIPAL FINDINGS: Presymptomatic SOD1(G93A) rats (60-65 days old) received spinal lumbar injections of hNSCs. After cell grafting, disease onset, disease progression and lifespan were analyzed. In separate symptomatic SOD1(G93A) rats, the presence and functional conductivity of descending motor tracts (corticospinal and rubrospinal) was analyzed by spinal surface recording electrodes after electrical stimulation of the motor cortex. Silver impregnation of lumbar spinal cord sections and descending motor axon counting in plastic spinal cord sections were used to validate morphologically the integrity of descending motor tracts. Grafting of hNSCs into the lumbar spinal cord of SOD1(G93A) rats protected α-motoneurons in the vicinity of grafted cells, provided transient functional improvement, but offered no protection to α-motoneuron pools distant from grafted lumbar segments. Analysis of motor-evoked potentials recorded from the thoracic spinal cord of symptomatic SOD1(G93A) rats showed a near complete loss of descending motor tract conduction, corresponding to a significant (50-65%) loss of large caliber descending motor axons. CONCLUSIONS/SIGNIFICANCE: These data demonstrate that in order to achieve a more clinically-adequate treatment, cell-replacement/gene therapy strategies will likely require both spinal and supraspinal targets.


Subject(s)
Amyotrophic Lateral Sclerosis/surgery , Neural Stem Cells/transplantation , Spinal Cord/surgery , Stem Cell Transplantation , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Electric Stimulation , Evoked Potentials, Motor , Humans , Mutation , Rats , Rats, Transgenic , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Synapses/physiology
7.
PLoS One ; 7(3): e32581, 2012.
Article in English | MEDLINE | ID: mdl-22403675

ABSTRACT

We report here in adult rat viral vector mediate-gene knockdown in the primary sensory neurons and the associated cellular and behavior consequences. Self-complementary adeno-associated virus serotype 5 (AAV5) was constructed to express green fluorescent protein (GFP) and a small interfering RNA (siRNA) targeting mammalian target of rapamycin (mTOR). The AAV vectors were injected via an intrathecal catheter. We observed profound GFP expression in lumbar DRG neurons beginning at 2-week post-injection. Of those neurons, over 85% were large to medium-diameter and co-labeled with NF200, a marker for myelinated fibers. Western blotting of mTOR revealed an 80% reduction in the lumbar DRGs (L4-L6) of rats treated with the active siRNA vectors compared to the control siRNA vector. Gene knockdown became apparent as early as 7-day post-injection and lasted for at least 5 weeks. Importantly, mTOR knockdown occurred in large (NF200) and small-diameter neurons (nociceptors). The viral administration induced an increase of Iba1 immunoreactivity in the DRGs, which was likely attributed to the expression of GFP but not siRNA. Rats with mTOR knockdown in DRG neurons showed normal general behavior and unaltered responses to noxious stimuli. In conclusion, intrathecal AAV5 is a highly efficient vehicle to deliver siRNA and generate gene knockdown in DRG neurons. This will be valuable for both basic research and clinic intervention of diseases involving primary sensory neurons.


Subject(s)
Dependovirus/genetics , Ganglia, Spinal/metabolism , Ganglia, Spinal/virology , Gene Knockdown Techniques/methods , Injections, Spinal , Animals , Ganglia, Spinal/cytology , Genetic Vectors/genetics , Genetic Vectors/toxicity , Male , Neurons/drug effects , Neurons/metabolism , Neurons/virology , Nociception , RNA, Small Interfering/genetics , RNA, Small Interfering/toxicity , Rats , Rats, Sprague-Dawley , TOR Serine-Threonine Kinases/deficiency , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Transduction, Genetic
8.
PLoS One ; 7(1): e30561, 2012.
Article in English | MEDLINE | ID: mdl-22291989

ABSTRACT

BACKGROUND: Loss of GABA-mediated pre-synaptic inhibition after spinal injury plays a key role in the progressive increase in spinal reflexes and the appearance of spasticity. Clinical studies show that the use of baclofen (GABA(B) receptor agonist), while effective in modulating spasticity is associated with major side effects such as general sedation and progressive tolerance development. The goal of the present study was to assess if a combined therapy composed of spinal segment-specific upregulation of GAD65 (glutamate decarboxylase) gene once combined with systemic treatment with tiagabine (GABA uptake inhibitor) will lead to an antispasticity effect and whether such an effect will only be present in GAD65 gene over-expressing spinal segments. METHODS/PRINCIPAL FINDINGS: Adult Sprague-Dawley (SD) rats were exposed to transient spinal ischemia (10 min) to induce muscle spasticity. Animals then received lumbar injection of HIV1-CMV-GAD65 lentivirus (LVs) targeting ventral α-motoneuronal pools. At 2-3 weeks after lentivirus delivery animals were treated systemically with tiagabine (4, 10, 20 or 40 mg/kg or vehicle) and the degree of spasticity response measured. In a separate experiment the expression of GAD65 gene after spinal parenchymal delivery of GAD65-lentivirus in naive minipigs was studied. Spastic SD rats receiving spinal injections of the GAD65 gene and treated with systemic tiagabine showed potent and tiagabine-dose-dependent alleviation of spasticity. Neither treatment alone (i.e., GAD65-LVs injection only or tiagabine treatment only) had any significant antispasticity effect nor had any detectable side effect. Measured antispasticity effect correlated with increase in spinal parenchymal GABA synthesis and was restricted to spinal segments overexpressing GAD65 gene. CONCLUSIONS/SIGNIFICANCE: These data show that treatment with orally bioavailable GABA-mimetic drugs if combined with spinal-segment-specific GAD65 gene overexpression can represent a novel and highly effective anti-spasticity treatment which is associated with minimal side effects and is restricted to GAD65-gene over-expressing spinal segments.


Subject(s)
GABA Agonists/therapeutic use , Genetic Therapy , Glutamate Decarboxylase/genetics , Muscle Spasticity/therapy , Spine/metabolism , Animals , Anticonvulsants/administration & dosage , Anticonvulsants/adverse effects , Anticonvulsants/therapeutic use , Cells, Cultured , Combined Modality Therapy , Embryo, Mammalian , Female , GABA Agonists/administration & dosage , GABA Agonists/adverse effects , Gene Expression Regulation/physiology , Genetic Therapy/methods , Glutamate Decarboxylase/administration & dosage , Glutamate Decarboxylase/adverse effects , Injections, Spinal , Male , Muscle Spasticity/drug therapy , Muscle Spasticity/genetics , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/adverse effects , Neuroprotective Agents/therapeutic use , Nipecotic Acids/administration & dosage , Nipecotic Acids/adverse effects , Nipecotic Acids/therapeutic use , Rats , Rats, Sprague-Dawley , Spine/pathology , Swine , Swine, Miniature , Tiagabine
9.
Nature ; 482(7384): 216-20, 2012 Jan 25.
Article in English | MEDLINE | ID: mdl-22278060

ABSTRACT

Our understanding of Alzheimer's disease pathogenesis is currently limited by difficulties in obtaining live neurons from patients and the inability to model the sporadic form of the disease. It may be possible to overcome these challenges by reprogramming primary cells from patients into induced pluripotent stem cells (iPSCs). Here we reprogrammed primary fibroblasts from two patients with familial Alzheimer's disease, both caused by a duplication of the amyloid-ß precursor protein gene (APP; termed APP(Dp)), two with sporadic Alzheimer's disease (termed sAD1, sAD2) and two non-demented control individuals into iPSC lines. Neurons from differentiated cultures were purified with fluorescence-activated cell sorting and characterized. Purified cultures contained more than 90% neurons, clustered with fetal brain messenger RNA samples by microarray criteria, and could form functional synaptic contacts. Virtually all cells exhibited normal electrophysiological activity. Relative to controls, iPSC-derived, purified neurons from the two APP(Dp) patients and patient sAD2 exhibited significantly higher levels of the pathological markers amyloid-ß(1-40), phospho-tau(Thr 231) and active glycogen synthase kinase-3ß (aGSK-3ß). Neurons from APP(Dp) and sAD2 patients also accumulated large RAB5-positive early endosomes compared to controls. Treatment of purified neurons with ß-secretase inhibitors, but not γ-secretase inhibitors, caused significant reductions in phospho-Tau(Thr 231) and aGSK-3ß levels. These results suggest a direct relationship between APP proteolytic processing, but not amyloid-ß, in GSK-3ß activation and tau phosphorylation in human neurons. Additionally, we observed that neurons with the genome of one sAD patient exhibited the phenotypes seen in familial Alzheimer's disease samples. More generally, we demonstrate that iPSC technology can be used to observe phenotypes relevant to Alzheimer's disease, even though it can take decades for overt disease to manifest in patients.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Neurons/metabolism , Aged, 80 and over , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Astrocytes/cytology , Biomarkers/metabolism , Cells, Cultured , Cellular Reprogramming , Coculture Techniques , Endosomes/metabolism , Enzyme Activation , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Glycogen Synthase Kinase 3/metabolism , Humans , Male , Middle Aged , Models, Biological , Neurons/drug effects , Neurons/pathology , Peptide Fragments/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protease Inhibitors/pharmacology , Proteolysis , Synapsins/metabolism , tau Proteins/metabolism
10.
Cell Transplant ; 20(8): 1153-61, 2011.
Article in English | MEDLINE | ID: mdl-21669047

ABSTRACT

Previous rodent studies employing monotherapy or combined immunosuppressive regimens have demonstrated a variable degree of spinal xenograft survival in several spinal neurodegenerative models including spinal ischemia, trauma, or amyotrophic lateral sclerosis (ALS). Accordingly, the characterization of optimal immunosuppressive protocols for the specific neurodegenerative model is critical to ensure reliable assessment of potential long-term therapeutic effects associated with cell replacement. In the present study we characterized the survival of human spinal stem cells when grafted into the lumbar spinal cords of a rodent model of ALS, SOD1 (G93A) male and female rats (60-67 days old). Four different immunosuppressive protocols were studied: i) FK506 (q12h); ii) FK506 (qd) + mycophenolate (PO; q12h, up to 7 days postop); iii) FK506 (qd) + mycophenolate (IP; q12h, up to 7 days postop); and iv) FK506 (qd) + mycophenolate (IP; qd, up to 7 days postop). Three weeks after cell grafting the number of surviving human cells was then systematically assessed. The highest density of grafted cells was seen in animals treated with FK506 (qd) and mycophenolate (IP; qd; an average 915 ± 95 grafted cells per spinal cord section). The majority of hNUMA-positive cells colocalized with doublecortin (DCX) immunoreactivity. DCX-positive neurons showed extensive axodendritic sprouting toward surrounding host neurons. In addition, migrating grafted cells were identified up to 500 µm from the graft. In animals treated with FK506 (q12h), FK506 (qd) + mycophenolate (PO; q12h) or FK506 (qd) + mycophenolate (IP; q12h), 11.8 ± 3.4%, 61.2 ± 7.8%, and 99.4 ± 8.9% [expressed as percent of the FK506 (qd) and mycophenolate (IP; qd)] cell survival was seen, respectively. In contrast to animals treated with a combination of FK506 + mycophenolate, robust CD4/8 immunoreactivity was identified in the vicinity of the injection tract in animals treated with FK506 only. These data suggest that a combined, systemically delivered immunosuppression regimen including FK506 and mycophenolate can significantly improve survival of human spinal stem cells after intraspinal transplantation in SOD1 (G93A) rats.


Subject(s)
Amyotrophic Lateral Sclerosis/therapy , Immunosuppression Therapy/methods , Immunosuppressive Agents/therapeutic use , Spinal Cord/cytology , Stem Cell Transplantation , Stem Cells/cytology , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/immunology , Animals , Cell Survival/drug effects , Disease Models, Animal , Doublecortin Domain Proteins , Doublecortin Protein , Female , Fluorescent Antibody Technique , Humans , Immune Tolerance/drug effects , Male , Microtubule-Associated Proteins/metabolism , Neuropeptides/metabolism , Rats , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Tacrolimus/pharmacology , Tacrolimus/therapeutic use
11.
Br J Pharmacol ; 161(5): 976-85, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20977450

ABSTRACT

BACKGROUND AND PURPOSE: Baclofen (a GABA(B) receptor agonist) is the most commonly used anti-spasticity agent in clinical practice. While effective when administered spinally or systemically, the development of progressive tolerance represents a serious limitation for its long-term use. The goal of the present study was to characterize the treatment potency after intrathecal or systemic treatment with the selective AMPA receptor antagonist NGX424 on stretch reflex activity (SRA) and background muscle activity (BMA) in rats with developed baclofen tolerance. EXPERIMENTAL APPROACH: Animals were exposed to 10 min of spinal ischaemia to induce an increase in BMA and SRA. Selected animals were implanted with an intrathecal PE-5 catheter and infused intrathecally with baclofen (1 µg·h⁻¹ ) for 14 days. Before and after baclofen infusion, changes in BMA and SRA were measured at 2 day intervals. After development of baclofen tolerance, the animals were injected intrathecally (1 µg) or subcutaneously (3, 6 or 12 mg·kg⁻¹) with NGX424, and changes in BMA and SRA were measured. KEY RESULTS: Intrathecal or systemic delivery of NGX424 significantly suppressed the BMA and SRA in baclofen-tolerant animals. This effect was dose dependent. The magnitude of BMA and SRA suppression seen after 1 µg (intrathecal) or 12 mg·kg ⁻¹ (s.c.) of NGX424 injection was similar to that seen during the first 5 days of baclofen infusion. CONCLUSIONS AND IMPLICATIONS These data demonstrate that the use of NGX424 can represent an effective therapy to modulate chronic spasticity in patients who are refractory or tolerant to baclofen treatment.


Subject(s)
Baclofen/pharmacology , Isoquinolines/pharmacology , Muscle Spasticity/drug therapy , Reflex, Stretch/drug effects , Tetrazoles/pharmacology , Animals , Baclofen/administration & dosage , Dose-Response Relationship, Drug , Drug Tolerance , GABA-B Receptor Agonists/administration & dosage , GABA-B Receptor Agonists/pharmacology , Injections, Spinal , Injections, Subcutaneous , Isoquinolines/administration & dosage , Male , Muscle Relaxants, Central/administration & dosage , Muscle Relaxants, Central/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, AMPA/antagonists & inhibitors , Tetrazoles/administration & dosage
12.
PLoS One ; 5(8): e12148, 2010 Aug 13.
Article in English | MEDLINE | ID: mdl-20730054

ABSTRACT

BACKGROUND: Due to the inherent sensitivity of human embryonic stem cells (hESCs) to manipulations, the recovery and survival of hESCs after fluorescence-activated cell sorting (FACS) can be low. Additionally, a well characterized and robust methodology for performing FACS on hESCs using multiple-cell surface markers has not been described. The p160-Rho-associated coiled kinase (ROCK) inhibitor, Y-27632, previously has been identified as enhancing survival of hESCs upon single-cell dissociation, as well as enhancing recovery from cryopreservation. Here we examined the application of Y-27632 to hESCs after FACS to improve survival in both feeder-dependent and feeder-independent growth conditions. METHODOLOGY/PRINCIPAL FINDINGS: HESCs were sorted using markers for SSEA-3, TRA-1-81, and SSEA-1. Cells were plated after sorting for 24 hours in either the presence or the absence of Y-27632. In both feeder-dependent and feeder-independent conditions, cell survival was greater when Y-27632 was applied to the hESCs after sort. Specifically, treatment of cells with Y-27632 improved post-sort recovery up to four fold. To determine the long-term effects of sorting with and without the application of Y-27632, hESCs were further analyzed. Specifically, hESCs sorted with and without the addition of Y-27632 retained normal morphology, expressed hESC-specific markers as measured by immunocytochemistry and flow cytometry, and maintained a stable karyotype. In addition, the hESCs could differentiate into three germ layers in vitro and in vivo in both feeder-dependent and feeder-independent growth conditions. CONCLUSIONS/SIGNIFICANCE: The application of Y-27632 to hESCs after cell sorting improves cell recovery with no observed effect on pluripotency, and enables the consistent recovery of hESCs by FACS using multiple surface markers. This improved methodology for cell sorting of hESCs will aid many applications such as removal of hESCs from secondary cell types, identification and isolation of stem cell subpopulations, and generation of single cell clones. Finally, these results demonstrate an additional application of ROCK inhibition to hESC research.


Subject(s)
Amides/pharmacology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Flow Cytometry/methods , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , rho-Associated Kinases/antagonists & inhibitors , Biomarkers/metabolism , Cell Culture Techniques , Cell Differentiation/drug effects , Embryonic Stem Cells/metabolism , Humans , Karyotyping
13.
Stem Cells Dev ; 19(12): 1983-93, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20406098

ABSTRACT

Most stem cell therapies involve direct, intraparachymal placement of neural progenitor cells. These cells provide physical support to the endogenous neuronal population and may be engineered to provide in situ growth factor support. Insulin-like growth factor-I (IGF-I) has potent neurotrophic and neuroprotective properties and is expressed by human neural stem cells (hNSCs). IGF-I is implicated in multiple aspects of cell behavior, including proliferation, differentiation, and survival. Enhancing hNSC function through IGF-I overexpression may increase the benefits of stem cell therapy. As a first step to that goal, we examined the direct effects of IGF-I on hNSC behavior in vitro. We demonstrate that IGF-I treatment enhances both the number and length of hNSC neurites. This is correlated with a decrease in proliferation, suggesting that IGF-I promotes neurite outgrowth but not proliferation. While IGF-I activates both AKT and MAPK signaling in hNSCs, we demonstrate that IGF-I-mediated neurite outgrowth is dependent only on AKT signaling. Finally, we demonstrate that IGF-I is neuroprotective after glutamate exposure in a model of excitotoxic cell death.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Neurites/metabolism , Neurogenesis , Spinal Cord/cytology , Blotting, Western , Cell Death/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Fluorescent Antibody Technique , Glutamic Acid/pharmacology , Humans , In Situ Nick-End Labeling , Mitogen-Activated Protein Kinases/metabolism , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Polymerase Chain Reaction , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
14.
Cell Transplant ; 19(9): 1103-22, 2010.
Article in English | MEDLINE | ID: mdl-20412634

ABSTRACT

In recent studies using a rat aortic balloon occlusion model, we have demonstrated that spinal grafting of rat or human neuronal precursors or human postmitotic hNT neurons leads to progressive amelioration of spasticity and rigidity and corresponding improvement in ambulatory function. In the present study, we characterized the optimal dosing regimen and safety profile of human spinal stem cells (HSSC) when grafted into the lumbar spinal cord segments of naive immunosuppressed minipigs. Gottingen-Minnesota minipigs (18-23 kg) were anesthetized with halothane, mounted into a spine-immobilization apparatus, and received five bilateral injections of HSSC delivered in 2, 4, 6, 8, or 10 µl of media targeted into L2-L5 central gray matter (lamina VII). The total number of delivered cells ranged between 2,500 and 100,000 per injection. Animals were immunosuppressed with Prograf® for the duration of study. After cell grafting, ambulatory function was monitored daily using a Tarlov's score. Sensory functions were assessed by mechanically evoked skin twitch test. Animals survived for 6-7 weeks. Three days before sacrifice animals received daily injections of bromodeoxyuridine (100 mg/kg; IV) and were then transcardially perfused with 4% paraformaldehyde. Th12-L6 spinal column was then dissected; the spinal cord was removed and scanned with MRI. Lumbar transverse spinal cord sections were then cut and stained with a combination of human-specific (hNUMA, hMOC, hNSE, hSYN) or nonspecific (DCX, MAP2, GABA, CHAT) antibodies. The total number of surviving cells was estimated using stereological quantification. During the first 12-24 h after cell grafting, a modest motor weakness was observed in three of eight animals but was no longer present at 4 days to 7 weeks. No sensory dysfunction was seen at any time point. Postmortem MRI scans revealed the presence of the individual grafts in the targeted spinal cord areas. Histological examination of spinal cord sections revealed the presence of hNUMA-immunoreactive grafted cells distributed between the base of the dorsal horn and the ventral horn. In all grafts intense hMOC, DCX, and hSYN immunoreactivity in grafted cells was seen. In addition, a rich axodendritic network of DCX-positive processes was identified extending 300-700 µm from the grafts. On average, 45% of hNUMA-positive neurons were GABA immunoreactive. Stereological analysis of hNUMA-positive cells showed an average of 2.5- to 3-fold increase in number of surviving cells compared with the number of injected cells. Analysis of spinal structural morphology showed that in animals injected with more than 50,000 cells/injection or volumes of injectate higher than 6 µl/injection there was tissue expansion and disruption of the local axodendritic network. Based on these data the safe total number of injected cells and volume of injectate were determined to be 30,000 cells delivered in ≤6 µl of media. These data demonstrate that highly reproducible delivery of a potential cell therapeutic candidate into spinal parenchyma can be achieved across a wide range of cell doses by direct intraspinal injections. The resulting grafts uniformly showed robust cell survival and progressive neuronal maturation.


Subject(s)
Graft Survival/physiology , Neural Stem Cells/transplantation , Spinal Cord/cytology , Stem Cell Transplantation/methods , Animals , Cell Growth Processes/physiology , Cell Survival/immunology , Cell Survival/physiology , Doublecortin Protein , Female , Graft Survival/immunology , Humans , Immunosuppression Therapy , Male , Mice , Neural Stem Cells/cytology , Reproducibility of Results , Spinal Cord/surgery , Swine , Swine, Miniature
15.
Growth Factors ; 27(3): 133-40, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19294549

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is characterized by loss of both upper and lower motor neurons. ALS progression is complex and likely due to cellular dysfunction at multiple levels, including mitochondrial dysfunction, glutamate excitotoxicity, oxidative stress, axonal dysfunction, reactive astrocytosis, and mutant superoxide dismutase expression, therefore, treatment must provide neuronal protection from multiple insults. A significant amount of ALS research focuses on growth factor-based therapies. Growth factors including insulin-like growth factor-I, vascular endothelial growth factor, brain-derived neurotrophic factor, and glial-derived neurotrophic factor exhibit robust neuroprotective effects on motor neurons in ALS models. Issues concerning growth factor delivery, stability and unwanted side effects slow the transfer of these treatments to human ALS patients. Stem cells represent a new therapeutic approach offering both cellular replacement and trophic support for the existing population. Combination therapy consisting of stem cells expressing beneficial growth factors may provide a comprehensive treatment for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Intercellular Signaling Peptides and Proteins/therapeutic use , Motor Neurons/cytology , Stem Cells/cytology , Amyotrophic Lateral Sclerosis/therapy , Axons/physiology , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Motor Neurons/physiology , Oxidative Stress , Stem Cell Transplantation , Stem Cells/physiology
16.
J Neurosci ; 29(4): 1093-104, 2009 Jan 28.
Article in English | MEDLINE | ID: mdl-19176818

ABSTRACT

Astrocytes in the CNS respond to tissue damage by becoming reactive. They migrate, undergo hypertrophy, and form a glial scar that inhibits axon regeneration. Therefore, limiting astrocytic responses represents a potential therapeutic strategy to improve functional recovery. It was recently shown that the epidermal growth factor (EGF) receptor is upregulated in astrocytes after injury and promotes their transformation into reactive astrocytes. Furthermore, EGF receptor inhibitors were shown to enhance axon regeneration in the injured optic nerve and promote recovery after spinal cord injury. However, the signaling pathways involved were not elucidated. Here we show that in cultures of adult spinal cord astrocytes EGF activates the mTOR pathway, a key regulator of astrocyte physiology. This occurs through Akt-mediated phosphorylation of the GTPase-activating protein Tuberin, which inhibits Tuberin's ability to inactivate the small GTPase Rheb. Indeed, we found that Rheb is required for EGF-dependent mTOR activation in spinal cord astrocytes, whereas the Ras-MAP kinase pathway does not appear to be involved. Moreover, astrocyte growth and EGF-dependent chemoattraction were inhibited by the mTOR-selective drug rapamycin. We also detected elevated levels of activated EGF receptor and mTOR signaling in reactive astrocytes in vivo in an ischemic model of spinal cord injury. Furthermore, increased Rheb expression likely contributes to mTOR activation in the injured spinal cord. Interestingly, injured rats treated with rapamycin showed reduced signs of reactive gliosis, suggesting that rapamycin could be used to harness astrocytic responses in the damaged nervous system to promote an environment more permissive to axon regeneration.


Subject(s)
Astrocytes/metabolism , Monomeric GTP-Binding Proteins/metabolism , Neuropeptides/metabolism , Protein Kinases/metabolism , Signal Transduction/physiology , Spinal Cord Injuries/pathology , Up-Regulation/physiology , Analysis of Variance , Animals , Astrocytes/drug effects , Cells, Cultured , Chromones/pharmacology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/drug effects , Epidermal Growth Factor/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Excitatory Amino Acid Transporter 2/genetics , Excitatory Amino Acid Transporter 2/metabolism , Flavonoids/pharmacology , Glial Fibrillary Acidic Protein/metabolism , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Male , Monomeric GTP-Binding Proteins/genetics , Morpholines/pharmacology , Neuropeptides/genetics , Protein Kinases/genetics , RNA, Messenger/metabolism , Ras Homolog Enriched in Brain Protein , Rats , Rats, Sprague-Dawley , Sirolimus/pharmacology , Sirolimus/therapeutic use , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , TOR Serine-Threonine Kinases , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection/methods , Up-Regulation/drug effects , Vimentin/genetics , Vimentin/metabolism
17.
J Neurosci Res ; 87(4): 948-55, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18855936

ABSTRACT

Activation of p38 mitogen-activated protein kinase (MAPK) in the spinal cord has been implicated in the development and maintenance of pain states. In this study, we tested whether p38 MAPK is involved in the response to first-degree burn of the hind paw. This injury induces central sensitization leading to tactile allodynia and is mediated by activation of Ca(2+) permeable AMPA/kainate receptors through PKC and PKA. We demonstrate that p38 MAPK is rapidly and robustly activated in the superficial spinal dorsal horn after mild thermal injury to the hind paw. Activated p38 MAPK was localized primarily to microglia and to a lesser extent in oligodendrocytes and lamina II neurons. Astrocytes were not involved in the p38 MAPK response. Intrathecal pretreatment of pharmacological inhibitors of p38 MAPK (SB203580, SD-282) dose-dependently blocked development of tactile allodynia, a characteristic of the first-degree burn model. The effects of the inhibitors on tactile allodynia were lost when they were administered after injury. These studies identify p38 MAPK as a major mediator of tactile allodynia, most likely activated downstream of AMPA/kainate receptors.


Subject(s)
Burns/physiopathology , Pain/enzymology , Spinal Cord/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Burns/enzymology , Disease Models, Animal , Enzyme Activation , Enzyme Inhibitors/administration & dosage , Imidazoles/administration & dosage , Indoles/administration & dosage , Male , Microglia/enzymology , Neurons/enzymology , Oligodendroglia/enzymology , Pain/drug therapy , Phosphorylation , Pyridines/administration & dosage , Rats , Rats, Sprague-Dawley
18.
J Neurosci ; 27(42): 11179-91, 2007 Oct 17.
Article in English | MEDLINE | ID: mdl-17942713

ABSTRACT

Using a rat model of ischemic paraplegia, we examined the expression of spinal AMPA receptors and their role in mediating spasticity and rigidity. Spinal ischemia was induced by transient occlusion of the descending aorta combined with systemic hypotension. Spasticity/rigidity were identified by simultaneous measurements of peripheral muscle resistance (PMR) and electromyography (EMG) before and during ankle flexion. In addition, Hoffman reflex (H-reflex) and motor evoked potentials (MEPs) were recorded from the gastrocnemius muscle. Animals were implanted with intrathecal catheters for drug delivery and injected with the AMPA receptor antagonist NGX424 (tezampanel), glutamate receptor 1 (GluR1) antisense, or vehicle. Where intrathecal vehicle had no effect, intrathecal NGX424 produced a dose-dependent suppression of PMR [ED50 of 0.44 microg (0.33-0.58)], as well as tonic and ankle flexion-evoked EMG activity. Similar suppression of MEP and H-reflex were also seen. Western blot analyses of lumbar spinal cord tissue from spastic animals showed a significant increase in GluR1 but decreased GluR2 and GluR4 proteins. Confocal and electron microscopic analyses of spinal cord sections from spastic animals revealed increased GluR1 immunoreactivity in reactive astrocytes. Selective GluR1 knockdown by intrathecal antisense treatment resulted in a potent reduction of spasticiy and rigidity and concurrent downregulation of neuronal/astrocytic GluR1 in the lumbar spinal cord. Treatment of rat astrocyte cultures with AMPA led to dose-dependent glutamate release, an effect blocked by NGX424. These data suggest that an AMPA/kainate receptor antagonist can represent a novel therapy in modulating spasticity/rigidity of spinal origin and that astrocytes may be a potential target for such treatment.


Subject(s)
Astrocytes/metabolism , Muscle Rigidity/metabolism , Muscle Spasticity/metabolism , Receptors, AMPA/biosynthesis , Receptors, AMPA/genetics , Spinal Cord Ischemia/metabolism , Animals , Astrocytes/cytology , Cells, Cultured , Gene Expression Regulation/physiology , Male , Muscle Rigidity/etiology , Muscle Rigidity/genetics , Muscle Spasticity/etiology , Muscle Spasticity/genetics , Rats , Rats, Sprague-Dawley , Receptors, AMPA/physiology , Spinal Cord/cytology , Spinal Cord/metabolism , Spinal Cord Ischemia/complications , Spinal Cord Ischemia/genetics
19.
Eur J Neurosci ; 26(7): 1721-37, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17897390

ABSTRACT

Amyotrophic lateral sclerosis (ALS), spinal bulbar muscular atrophy (or Kennedy's disease), spinal muscular atrophy and spinal muscular atrophy with respiratory distress 1 are neurodegenerative disorders mainly affecting motor neurons and which currently lack effective therapies. Recent studies in animal models as well as primary and embryonic stem cell models of ALS, utilizing over-expression of mutated forms of Cu/Zn superoxide dismutase 1, have shown that motor neuron degeneration in these models is in part a non cell-autonomous event and that by providing genetically non-compromised supporting cells such as microglia or growth factor-excreting cells, onset can be delayed and survival increased. Using models of acute motor neuron injury it has been shown that embryonic stem cell-derived motor neurons implanted into the spinal cord can innervate muscle targets and improve functional recovery. Thus, a rationale exists for the development of cell therapies in motor neuron diseases aimed at either protecting and/or replacing lost motor neurons, interneurons as well as non-neuronal cells. This review evaluates approaches used in animal models of motor neuron disorders and their therapeutic relevance.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Motor Neuron Disease/therapy , Stem Cells/physiology , Animals , Disease Models, Animal , Humans
20.
J Neurosci Methods ; 161(1): 23-31, 2007 Mar 30.
Article in English | MEDLINE | ID: mdl-17083983

ABSTRACT

Substance P release from nociceptive primary afferents activates post-synaptic neurokinin-1 (NK-1) receptors causing subsequent NK-1 receptor internalization. Fluorescent immunohistochemistry is typically used to quantify NK-1 receptor internalization, an indirect measure of substance P (SP) release. However, this technique entails several limitations that restrict its application. Using simple subcellular fractionation and immunoblotting methods, we demonstrate that intrathecal SP invokes a rapid and dose-dependent increase in dorsal horn cytoplasmic NK-1 receptors. We also show that hind paw compression and noxious thermal stimulation increase cytoplasmic NK-1 receptor, when compared to sham stimulations. Fluorescent immunohistochemistry confirmed that increases in cytoplasmic NK-1 corresponded with increased NK-1 receptor internalization. Herein, we report that low-speed centrifugation and Western immunoblotting provide NK-1 internalization results consistent with those obtained by more traditional methods. These data support previous findings demonstrating a role for spinal NK-1 receptors in nociceptive processing.


Subject(s)
Cell Fractionation/methods , Hyperalgesia/metabolism , Receptors, Neurokinin-1/metabolism , Spinal Cord/metabolism , Animals , Dose-Response Relationship, Drug , Functional Laterality , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Male , Protein Transport/drug effects , Protein Transport/physiology , Rats , Spinal Cord/ultrastructure , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Substance P
SELECTION OF CITATIONS
SEARCH DETAIL
...