Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Brain Res ; 1530: 1-12, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-23891717

ABSTRACT

An increasing number of in vitro and in vivo studies suggest that anesthesia and surgery could be risk factors for later cognitive impairment in the young and aged brain. General anesthesia has been shown to impair spatial memory in rats and this performance is dependent on hippocampal function and postnatal hippocampal neurogenesis. Anesthetic induced alteration of one or more stages of postnatal hippocampal neurogenesis may in part explain this cognitive impairment following anesthesia. Three different populations of proliferating cells in the dentate gyrus (DG) were labeled with different thymidine analogs (EdU, IdU, and CldU) at 4, 8, and 21 days, respectively, in young (3-month-old) and aged (20-month-old) rats prior to a 3h exposure to isoflurane, control, propofol, or 10% intralipid. 24h following general anesthesia, brains were collected for analysis. The number of cells co-localized with neuronal differentiation and maturation labels with each of the thymidine analogs was quantified. In addition, new cell proliferation 24hr following anesthesia was assessed with anti-Ki67. The effect of anesthesia on astrocytes was also assessed with anti-S100ß. Isoflurane or propofol did not affect new cell proliferation, as assessed by Ki67, in the DG of young or aged rats. However, propofol significantly decreased the number of differentiating neurons and increased the number of astrocytes in the DG of young, but not aged, rats. Isoflurane significantly decreased the number of maturing neurons and increased the number of astrocytes in the DG of aged, but not young, rats. Isoflurane and propofol anesthesia altered postnatal hippocampal neurogenesis in an age and agent dependent matter.


Subject(s)
Anesthetics, Inhalation/toxicity , Hippocampus/drug effects , Isoflurane/toxicity , Neurogenesis/drug effects , Neurons/drug effects , Propofol/toxicity , Aging/physiology , Anesthesia, General , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Dentate Gyrus/drug effects , Hippocampus/cytology , Male , Memory/drug effects , Memory/physiology , Neurogenesis/physiology , Neurons/cytology , Rats , Rats, Inbred F344
2.
Brain Res ; 1469: 114-28, 2012 Aug 21.
Article in English | MEDLINE | ID: mdl-22750125

ABSTRACT

Vascular pathology, including blood-brain/spinal cord barrier (BBB/BSCB) alterations, has recently been recognized as a key factor possibly aggravating motor neuron damage, identifying a neurovascular disease signature for ALS. However, BBB/BSCB competence in sporadic ALS (SALS) is still undetermined. In this study, BBB/BSCB integrity in postmortem gray and white matter of medulla and spinal cord tissue from SALS patients and controls was investigated. Major findings include (1) endothelial cell damage and pericyte degeneration, (2) severe intra- and extracellular edema, (3) reduced CD31 and CD105 expressions in endothelium, (4) significant accumulation of perivascular collagen IV, and fibrin deposits (5) significantly increased microvascular density in lumbar spinal cord, (6) IgG microvascular leakage, (7) reduced tight junction and adhesion protein expressions. Microvascular barrier abnormalities determined in gray and white matter of the medulla, cervical, and lumbar spinal cord of SALS patients are novel findings. Pervasive barrier damage discovered in ALS may have implications for disease pathogenesis and progression, as well as for uncovering novel therapeutic targets.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Blood-Brain Barrier/pathology , Medulla Oblongata/pathology , Spinal Cord/pathology , Aged , Aged, 80 and over , Amyotrophic Lateral Sclerosis/diagnostic imaging , Blood-Brain Barrier/diagnostic imaging , Disease Progression , Endothelial Cells/pathology , Endothelial Cells/ultrastructure , Female , Humans , Male , Medulla Oblongata/diagnostic imaging , Middle Aged , Spinal Cord/diagnostic imaging , Tight Junctions/pathology , Tight Junctions/ultrastructure , Ultrasonography
3.
Brain Res ; 1441: 38-46, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22297171

ABSTRACT

There is a growing body of evidence showing that a statistically significant number of people experience long-term changes in cognition after anesthesia. We hypothesize that this cognitive impairment may result from an anesthetic-induced alteration of postnatal hippocampal cell proliferation. To test this hypothesis, we investigated the effects of isoflurane and propofol on new cell proliferation and cognition of young (4 month-old) and aged (21 month-old). All rats were injected intraperitoneally (IP) with 50 mg/kg of 5-bromo-2-deoxyuridine (BrdU) immediately after anesthesia. A novel appetitive olfactory learning test was used to assess learning and memory two days after anesthesia. One week after anesthesia, rats were euthanized and the brains analyzed for new cell proliferation in the dentate gyrus, and proliferation and migration of newly formed cells in the subventricular zone to the olfactory bulb. We found that exposure to either isoflurane (p=0.017) or propofol (p=0.006) decreased hippocampal cell proliferation in young, but not in aged rats. This anesthetic-induced decrease was specific to new cell proliferation in the hippocampus, as new cell proliferation and migration to the olfactory bulb was unaffected. Isoflurane anesthesia produced learning impairment in aged rats (p=0.044), but not in young rats. Conversely, propofol anesthesia resulted in learning impairment in young (p=0.01), but not in aged rats. These results indicate that isoflurane and propofol anesthesia affect postnatal hippocampal cell proliferation and learning in an age dependent manner.


Subject(s)
Aging/drug effects , Cell Proliferation/drug effects , Dentate Gyrus/drug effects , Isoflurane/administration & dosage , Learning/drug effects , Propofol/administration & dosage , Aging/physiology , Anesthesia, Inhalation/methods , Anesthesia, Intravenous/methods , Animals , Dentate Gyrus/cytology , Dentate Gyrus/physiology , Learning/physiology , Male , Random Allocation , Rats , Rats, Inbred F344
4.
Stem Cell Rev Rep ; 6(4): 523-31, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20680520

ABSTRACT

Cerebral ischemia induces death of all neural cell types within the region affected by the loss of blood flow. We have shown that administering human umbilical cord blood cells after a middle cerebral artery occlusion in rats significantly reduces infarct size, presumably by rescuing cells within the penumbra. In this study we examined whether the cord blood cells enhanced astrocyte survival in an in vitro model of hypoxia with reduced glucose availability. Primary astrocyte cultures were incubated for 2 h in no oxygen (95% N, 5% CO(2)) and low glucose (1% compared to 4.5%) media. Cord blood mononuclear cells were added to half the cultures at the beginning of hypoxia. Astrocyte viability was determined using fluorescein diacetate/propidium iodide (FDA/PI) labeling and cytokine production by the astrocytes measured using ELISA. In some studies, T cells, B cells or monocytes/macrophages isolated from the cord blood mononuclear fraction with magnetic antibody cell sorting (MACS) were used instead to determine which cellular component of the cord blood mononuclear fraction was responsible for the observed effects. Co-culturing mononuclear cord blood cells with astrocytes during hypoxia stimulated production of IL-6 and IL-10 during hypoxia. The cord blood T cells decreased survival of the astrocytes after hypoxia but had no effect on the examined cytokines. Our data demonstrate that the tested cord blood fractions do not enhance astrocyte survival when delivered individually, suggesting there is either another cellular component that is neuroprotective or an interaction of all the cells is essential for protection.


Subject(s)
Astrocytes/cytology , Brain Ischemia/metabolism , Cytokines/metabolism , Fetal Blood/cytology , Animals , Astrocytes/metabolism , Cell Survival/physiology , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans , Hypoxia/metabolism , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Rats
5.
Stem Cells Dev ; 19(2): 221-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19788371

ABSTRACT

When human umbilical cord blood (HUCB) cells are systemically administered following middle cerebral artery occlusion (MCAO) in rats, they produce a reduction in infarct size resulting in recovery of motor function. Rats receiving HUCB cells have a less severe inflammatory response compared to MCAO stroke rats. The purpose of this study was to determine the interaction between HUCB cells and the main resident immune cells of the brain (microglia) under normoxic and hypoxic conditions in vitro. Primary microglial cultures were incubated for 2 h in no oxygen (95% N, 5% CO(2)) and low glucose (1%) media. Mononuclear HUCB cells were added to half the cultures at the beginning of the hypoxia conditions. Microglial viability was determined using fluorescein diacetate/propidium iodide (FDA/PI) labeling and cytokine expression using ELISA. In some studies, CD11b+ or CD19+ cells isolated from the HUCB mononuclear fraction with magnetic antibody cell sorting (MACS) were used instead of the mononuclear fraction. Co-culturing mononuclear HUCB cells with microglia decreased viability of the microglia during hypoxia. In the microglial monocultures, hypoxia significantly increased release of IL-1beta compared to normoxia, while adding HUCB cells in the hypoxia condition decreased IL-1beta concentrations to the same level as in the normoxia monocultures. Both CD11b+ and CD19+ HUCB cells decreased microglial viability during normoxia and hypoxia. Our data suggest that HUCB cells may produce a soluble factor that decreases viability of microglia.


Subject(s)
Cell Communication , Fetal Blood/cytology , Microglia/cytology , Animals , Antigens, CD19/metabolism , Brain , CD11b Antigen/metabolism , Cell Hypoxia , Cell Survival/drug effects , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Fetal Blood/metabolism , Fetus , Flow Cytometry , Glucose/pharmacology , Humans , Microglia/metabolism , Rats , Rats, Sprague-Dawley
6.
Pediatr Diabetes ; 9(6): 531-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19067891

ABSTRACT

BACKGROUND/OBJECTIVE: We previously reported that chronic hyperglycemia, but not hypoglycemia, was associated with the reduction of neuronal size in the rat brain. We hypothesized that hyperglycemia-induced changes in neuronal structure would have negative consequences, such as impaired learning and memory. We therefore assessed the effects of hyperglycemia and hypoglycemia on neuronal dendritic structure and cognitive functioning in young rats. DESIGN/METHODS: Experimental manipulations were conducted on male Wistar rats for 8 wk, beginning at 4 wk of age. At the completion of the treatments, all rats were trained in the radial-arm water maze, a spatial (hippocampus-dependent) learning and memory task. Three groups of rats were tested: an untreated control group, a streptozotocin-induced diabetic (STZ-D) group, and an intermittent hypoglycemic group. Following behavioral training, the brains of all animals were examined with histologic and biochemical measurements. RESULTS: Peripheral hyperglycemia was associated with significant increases in brain sorbitol (7.5 +/- 1.6 vs. 5.84 +/- 1.0 microM/mg) and inositol (9.6 +/- 1.4 vs. 7.1 +/- 1.1 microM/mg) and reduced taurine (0.65 +/- 0.1 vs. 1.3 +/- 0.1 mg/mg). Histologic evaluation revealed neurons with reduced dendritic branching and spine density in STZ-D rats but not in control or hypoglycemic animals. In addition, the STZ-D group exhibited impaired performance on the water maze memory test. CONCLUSIONS: Hyperglycemia, but not hypoglycemia, was associated with adverse effects on the brain polyol pathway activity, neuronal structural changes, and impaired long-term spatial memory. This finding suggests that the hyperglycemic component of diabetes mellitus has a greater adverse effect on brain functioning than does intermittent hypoglycemia.


Subject(s)
Dendrites/pathology , Diabetes Mellitus, Experimental/physiopathology , Hyperglycemia/complications , Memory Disorders/etiology , Neurons/pathology , Animals , Brain Chemistry/physiology , Cerebral Cortex/metabolism , Dendritic Spines/pathology , Dendritic Spines/physiology , Hippocampus/metabolism , Hyperglycemia/physiopathology , Hypoglycemia/complications , Hypoglycemia/physiopathology , Inositol/metabolism , Maze Learning/physiology , Memory/physiology , Rats , Rats, Wistar , Sorbitol/metabolism , Spatial Behavior/physiology , Taurine/metabolism
7.
Amyotroph Lateral Scler ; 9(6): 375-6, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18608097

ABSTRACT

Our letter to the editor addresses important questions regarding the role of the blood-spinal cord barrier in amyotrophic lateral sclerosis. The novel finding of barrier dysfunction in ALS has implications for disease pathogenesis. This discussion should prove of widespread interest to researchers and may help in formulating various new therapeutic strategies to protect barrier function and thus extend functionality and lifespan in ALS patients.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , Blood-Brain Barrier/physiopathology , Amyotrophic Lateral Sclerosis/genetics , Humans
8.
Neurotox Res ; 13(3-4): 241-6, 2008.
Article in English | MEDLINE | ID: mdl-18522903

ABSTRACT

The 796RMB cell line is a multipotent stem cell line isolated from human fetal midbrain tissues, a region from which dopamine neurons of the substantia nigra develop. It would be useful to increase the dopaminergic characteristics of this cell line to enhance its usefulness as a cell therapy for Parkinson's disease utilizing transplantation protocols. Sertoli cells and its conditioned media isolated from the testis have been previously shown to enhance tyrosine hydroxylase expression in ventral mesencephalon neurons both in vitro and in vivo. Therefore, the present preliminary study investigated the ability of Sertoli cell pre-conditioned medium to enhance differentiation of the 796MB cell line toward the domaminergic phenotype. Results showed that secretory products derived from Sertoli cell conditioned medium increased cell proliferation and enhanced dopaminergic neuronal differentiation of the 796RMB cell line. These findings may lead to alternative therapeutic cell transplantation protocols for the treatment of Parkinson's disease.


Subject(s)
Culture Media, Conditioned/pharmacology , Mesencephalon/cytology , Neurons/physiology , Sertoli Cells/chemistry , Stem Cells/drug effects , Animals , Animals, Newborn , Cell Count/methods , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dopamine/metabolism , Fetus , Humans , Male , Microtubule-Associated Proteins/metabolism , Rats , Tyrosine 3-Monooxygenase/metabolism
9.
Curr Neurovasc Res ; 5(2): 118-24, 2008 May.
Article in English | MEDLINE | ID: mdl-18473828

ABSTRACT

Monocyte chemoattractant protein-1 (MCP-1) and macrophage inflammatory protein (MIP-1alpha) are implicated in monocyte infiltration into the central nervous system (CNS) under pathological conditions. We previously showed that in vivo human umbilical cord blood cells (HUCB) migrate toward brain injury after middle cerebral artery occlusion (MCAO). We hypothesized that MCP-1 and MIP-1alpha may participate in the recruitment of HUCB towards the injury. Sprague-Dawley rats were subjected to middle cerebral artery occlusion (MCAO), and 24 hours later the production of MCP-1 and MIP-1alpha in the brain was examined with immunohistochemistry, ELISA, and western blotting. The chemotactic effect of MCP-1 and MIP-1alpha, and the expression of MCP-1 receptor CCR2 and MIP-1alpha receptor CCR1, CCR5 on the surface of HUCB were also examined. MCP-1 and MIP-1alpha expression were significantly increased in the ischemic hemisphere of brain, and significantly promoted HUCB cell migration compared to the contralateral side. This cell migration was neutralized with polyclonal antibodies against MCP-1 or MIP-1alpha. Also chemokine receptors were constitutively expressed on the surface of HUCB cells. The data suggested that the increased chemokines in the ischemic area can bind cell surface receptors on HUCB, and induce cell infiltration of systemically delivered HUCB cells into the CNS in vivo.


Subject(s)
Chemokine CCL2/therapeutic use , Chemokine CCL3/therapeutic use , Fetal Blood/cytology , Stroke/drug therapy , Stroke/surgery , Animals , Brain/cytology , Cell Movement , Cells, Cultured , Chemokine CCL2/metabolism , Chemokine CCL3/metabolism , Disease Models, Animal , Embryo, Mammalian , Humans , Neuroglia , Neurons , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Chemokine , Tubulin/metabolism
10.
Methods Mol Biol ; 438: 383-401, 2008.
Article in English | MEDLINE | ID: mdl-18369773

ABSTRACT

Stem cell transplantation to replace damaged tissue or correct metabolic disease holds the promise of helping a myriad of human afflictions. Although a great deal of attention has focused on pluripotent stem cells derived from embryos, adult stem cells have been described in a variety of tissues, and they likely will prove to be as beneficial as embryonic stem cells in cell replacement therapy and control of inbred errors of metabolism. We describe methods by which stem cells can be introduced into the nervous system, although the techniques are applicable to any portion of the body to be targeted or any cell that may be used for cell therapy. The first and most straight-forward method is introduction of stem cells directly into the brain parenchyma. The second, which in our hands has proven to be superior in some instances, is introduction of the stem cells into the circulatory system.


Subject(s)
Stem Cell Transplantation/methods , Stem Cells/metabolism , Animals , Mice , Postoperative Care , Rats
11.
PLoS One ; 2(11): e1205, 2007 Nov 21.
Article in English | MEDLINE | ID: mdl-18030339

ABSTRACT

BACKGROUND: The blood-brain barrier (BBB), blood-spinal cord barrier (BSCB), and blood-cerebrospinal fluid barrier (BCSFB) control cerebral/spinal cord homeostasis by selective transport of molecules and cells from the systemic compartment. In the spinal cord and brain of both ALS patients and animal models, infiltration of T-cell lymphocytes, monocyte-derived macrophages and dendritic cells, and IgG deposits have been observed that may have a critical role in motor neuron damage. Additionally, increased levels of albumin and IgG have been found in the cerebrospinal fluid in ALS patients. These findings suggest altered barrier permeability in ALS. Recently, we showed disruption of the BBB and BSCB in areas of motor neuron degeneration in the brain and spinal cord in G93A SOD1 mice modeling ALS at both early and late stages of disease using electron microscopy. Examination of capillary ultrastructure revealed endothelial cell degeneration, which, along with astrocyte alteration, compromised the BBB and BSCB. However, the effect of these alterations upon barrier function in ALS is still unclear. The aim of this study was to determine the functional competence of the BSCB in G93A mice at different stages of disease. METHODOLOGY/PRINCIPAL FINDINGS: Evans Blue (EB) dye was intravenously injected into ALS mice at early or late stage disease. Vascular leakage and the condition of basement membranes, endothelial cells, and astrocytes were investigated in cervical and lumbar spinal cords using immunohistochemistry. Results showed EB leakage in spinal cord microvessels from all G93A mice, indicating dysfunction in endothelia and basement membranes and confirming our previous ultrastructural findings on BSCB disruption. Additionally, downregulation of Glut-1 and CD146 expressions in the endothelial cells of the BSCB were found which may relate to vascular leakage. CONCLUSIONS/SIGNIFICANCE: Results suggest that the BSCB is compromised in areas of motor neuron degeneration in ALS mice at both early and late stages of the disease.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , Blood , Disease Models, Animal , Spinal Cord/metabolism , Superoxide Dismutase/metabolism , Amyotrophic Lateral Sclerosis/blood , Amyotrophic Lateral Sclerosis/cerebrospinal fluid , Animals , Disease Progression , Fluorescent Dyes , Immunohistochemistry , Mice , Motor Neurons/pathology , Superoxide Dismutase-1
12.
Brain Res ; 1157: 126-37, 2007 Jul 09.
Article in English | MEDLINE | ID: mdl-17512910

ABSTRACT

The purpose of this study was to determine the ultrastructure of the blood-brain barrier (BBB) and blood-spinal cord barrier (BSCB) in G93A SOD1 mice modeling ALS at different stages of disease. Electron microscope examination of brainstem, cervical and lumbar spinal cords was performed in ALS mice at early and late stages of disease. Our results show disorganized mitochondrial cristae and degenerating mitochondria in endothelial cells and neuropil, swollen astrocyte foot processes, swollen and degenerating capillary endothelial cells, astrocytes and motor neurons and extensive extracellular edema. In spite of progressive extracellular edema in neural tissue, capillary endothelial cell tight junctions appeared to remain intact in early and late symptomatic animals. Results show that disruption of BBB and BSCB was evident in areas of motor neuron degeneration in G93A mice at both early and late stages of disease. Capillary rupture was observed in brainstem in early symptomatic G93A mice. Capillary ultrastructure revealed that endothelial cell membrane and/or basement membrane damage occurred, followed by vascular leakage.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Blood-Brain Barrier/pathology , Brain Edema/pathology , Capillaries/pathology , Endothelial Cells/pathology , Superoxide Dismutase/genetics , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Astrocytes/pathology , Astrocytes/ultrastructure , Basement Membrane/pathology , Basement Membrane/ultrastructure , Blood-Brain Barrier/physiopathology , Blood-Brain Barrier/ultrastructure , Brain/blood supply , Brain/pathology , Brain/ultrastructure , Brain Edema/etiology , Brain Edema/physiopathology , Capillaries/physiopathology , Capillaries/ultrastructure , Disease Models, Animal , Endothelial Cells/ultrastructure , Extracellular Space/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , Mitochondria/pathology , Mitochondria/ultrastructure , Motor Neurons/pathology , Motor Neurons/ultrastructure , Mutation/genetics , Spinal Cord/blood supply , Spinal Cord/pathology , Spinal Cord/ultrastructure , Superoxide Dismutase-1 , Tight Junctions/pathology , Tight Junctions/ultrastructure
13.
Amyotroph Lateral Scler ; 7(4): 221-6, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17127560

ABSTRACT

hNT cells, derived from a human teratocarcinoma cell line, are versatile neuron-like cells that have been studied as possible treatment vehicles for neurodegenerative diseases. Previously, we showed the postponement of motor deficit symptoms in a G93A mouse model of amyotrophic lateral sclerosis (ALS) by transplanting hNT cells into the lumbar spinal cord. In this study, we examined the engraftment of hNT cells at multiple sites within the lumbar spinal cord by morphological analysis of neuritic process development. Results demonstrated that cells implanted at multiple sites established neuritic processes of different lengths independent of the number of cell implants. The hNT fiber outgrowth was a maximum of 0.15-0.3 mm from the transplants and mostly spread within the gray matter; interconnections between implants were not found. Therefore, we suggest that the observed postponement of motor deficit symptoms in G93A mice was not a result of neuritic outgrowth from the implanted hNT cells.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/surgery , Disease Models, Animal , Nerve Regeneration , Neurons/transplantation , Spinal Cord/pathology , Spinal Cord/surgery , Amyotrophic Lateral Sclerosis/congenital , Animals , Mice , Mice, Transgenic , Neurons/pathology , Reoperation , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Survival Rate , Treatment Outcome
14.
Prog Brain Res ; 157: 207-22, 2006.
Article in English | MEDLINE | ID: mdl-17046673

ABSTRACT

Numerous reports elucidate that tissue-specific stem cells are phenotypically plastic and their differentiation pathways are not strictly delineated. Although the identity of all the epigenetic factors which may trigger stem cells to make a lineage selection are still unknown, the plasticity of adult stem cells opens new approaches for their application in the treatment of various disorders. There is increasing researcher interest in hematopoietic stem cells for treatment of not only blood-related diseases but also various unrelated disorders including neurodegenerative diseases. Human umbilical cord blood (hUCB) cells, due to their primitive nature and ability to develop into nonhematopoietic cells of various tissue lineages, including neural cells, may be useful as an alternative cell source for cell-based therapies requiring either the replacement of individual cell types and/or substitution of missing substances. Here we focus on recent findings showing the robustness of adult stem cells derived from hUCB and their potential as a source of transplant cells for the treatment of diseased or injured brains and spinal cords. Depending upon the pathological microenvironment in which the hUCB cells are introduced, neuroprotective and/or trophic effects of these cells, from release of various growth or anti-inflammatory factors to moderation of immune-inflammatory effectors, may be more likely than neural replacement. These protective effects may prove essential to maintaining restored tissue integrity over the course of various diseases or injuries.


Subject(s)
Brain Injuries/therapy , Cell- and Tissue-Based Therapy , Spinal Cord Injuries/therapy , Amyotrophic Lateral Sclerosis/therapy , Animals , Fetal Blood/cytology , Humans , Stroke/therapy
15.
Stem Cells Dev ; 15(4): 497-506, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16978054

ABSTRACT

Our previous studies demonstrate enhanced neural protective effects of cord blood (CB) cells in comparison to stem cells from adult marrow. To determine further whether mesenchymal stem cells (MSCs) derived from human umbilical cord blood (hUCB) possess optimal characteristics for neural therapy, we isolated populations of plastic-adherent CB MSCs. These cells generated CD34-, CD45-, CD11b-, CD3-, CD19- cells in culture and failed to produce CFU-M, CFU-GEMM, or CFU-GM hematopoietic colonies in methylcellulose. However, cultured CB MSCs possessed a remarkable ability to support proliferation as well as differentiation of hematopoietic cells in vitro. In addition, supernatants from cultured CB MSCs promoted survival of NT2 N neural cells and peripheral blood mononuclear cells (MNCs) cultured under conditions designed to induce cell stress and limit protein synthesis. After incubation in neural differentiation medium, CB MSCs expressed the neural cell-surface antigen A2B5, the neurofilament polypeptide NF200, the oligodendrocyte precursor marker 04, intermediate filament proteins characteristic of neural differentiation (nestin and vimentin), as well as the astrocyte marker glial fibrillary acidic protein (GFAP) and the neural progenitor marker TUJ-1. We examined the immunomodulatory effects of the CB MSCs after co-culture with murine splenocytes. Whereas spleen cells from normal C57Bl/6 mice exhibited a prominent immunoglobulin M (IgM) response after immunization with the T cell-dependent antigen sheep red blood cells, this response was significantly decreased after incubation with CB MSCs. These data indicate that CB MSCs possess multiple utilities that may contribute to their therapeutic potency in the treatment of neurological disorders.


Subject(s)
Cell- and Tissue-Based Therapy , Fetal Blood/cytology , Mesenchymal Stem Cells/cytology , Nervous System Diseases/therapy , Animals , Biomarkers , Cell Differentiation , Cell Separation , Cell Survival , Cells, Cultured , Erythrocytes/immunology , Growth Substances/biosynthesis , Hematopoiesis/physiology , Humans , Immunoglobulin M/immunology , Nervous System Diseases/pathology , Neuroglia/cytology , Neurons/cytology , Sheep
16.
Brain Res ; 1096(1): 1-10, 2006 Jun 22.
Article in English | MEDLINE | ID: mdl-16780819

ABSTRACT

Sertoli cells (SCs) are testis-derived cells that secrete trophic factors important for the development of germ cells. Both porcine and rat SCs have been used as graft facilitators - neonatal porcine SCs to support islets in diabetes and 15-day-old rat SCs to enhance dopaminergic neuron transplants in Parkinson's disease models. However, there has never been a study examining the optimal SCs preparation to enhance tyrosine hydroxylase expression in the ventral mesencephalon (VM) neuron. The aim of this study was to compare the ability of both rat and porcine SCs to enhance tyrosine hydroxylase expression (TH) and neuronal survival at the same postnatal developmental ages. The SCs were isolated from 1-, 9-, or 15-day-old rat, or neonate (2-5 days), 2-month, or 4-month-old pig, and co-cultured with VM tissue from 13.5-day-old embryos. Our results showed that VM neurons co-cultured with SCs dispersed over the culture plate and had extensive neuritic outgrowth, while VM neurons cultured alone tended to cluster together forming a mass of cells with limited neurite outgrowth. TH expression was significantly increased when VM neurons were co-cultured with 15-day rat SCs or 2-month pig SCs but not when the cells were co-cultured with other ages of SCs. This suggests that secretion of trophic factors by SCs varies according to the developmental age, and it is critical for the success of graft facilitation that SCs from the appropriate age and species be used.


Subject(s)
Mesencephalon/cytology , Mesencephalon/enzymology , Neurons/enzymology , Sertoli Cells/physiology , Tyrosine 3-Monooxygenase/biosynthesis , Tyrosine 3-Monooxygenase/genetics , Animals , Animals, Newborn , Cell Division/physiology , Cell Separation , Cell Survival , Cells, Cultured , Coculture Techniques , Immunohistochemistry , Male , Mesencephalon/embryology , Neurites/physiology , Neurons/ultrastructure , Rats , Sertoli Cells/ultrastructure , Swine
17.
Brain Res ; 1076(1): 9-15, 2006 Mar 03.
Article in English | MEDLINE | ID: mdl-16487498

ABSTRACT

Children with diabetes onset before 5 years of age have reduced neurocognitive function. This problem has been attributed to hypoglycemia, a complication of insulin therapy. The eye, kidney, and nerve complications of diabetes (hyperglycemia) have been reduced by intensified insulin therapy which is associated with a 3-fold increase in severe hypoglycemia and therefore is not recommended for children less than 13 years of age. Since hyperglycemia is much more common than intermittent hypoglycemia during early childhood diabetes, it is important to determine if hyperglycemia affects brain growth and development. Rats were exposed to 4 weeks of either continuous hyperglycemia (diabetes) or intermittent (3 h, 3 times/week) hypoglycemia from 4 to 8 weeks of age. The brains of these animals were compared to those of similarly aged normal control animals. The cell number was increased, and the cell size reduced in the cortex of diabetic animals as assessed by DNA/wet weight of brain and protein/DNA content. Reduced amounts of protein, fatty acids, and cholesterol/microgram DNA also indicate smaller cells with reduced myelin content in the cortex of the diabetic animals. Histologic evaluation of these brains confirmed the biochemical findings. These observations require further confirmation and evaluation but indicate that continuous hyperglycemia may be more damaging than intermittent hypoglycemia to the developing brain. This is an important consideration for the management of diabetes mellitus in young children.


Subject(s)
Brain Injuries/complications , Brain Injuries/pathology , Hyperglycemia/etiology , Animals , Astrocytes/pathology , Cell Count/methods , Cerebral Cortex/pathology , Diabetes Mellitus, Experimental/complications , Glucose/metabolism , Hippocampus/pathology , Immunohistochemistry/methods , Male , Nerve Growth Factors/metabolism , Neurons/pathology , Neurons/physiology , Rats , Rats, Wistar , S100 Calcium Binding Protein beta Subunit , S100 Proteins/metabolism , Time Factors
18.
FASEB J ; 20(3): 485-7, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16401642

ABSTRACT

Numerous data support passage of maternal cells into the fetus during pregnancy in both human and animal models. However, functional benefits of maternal microchimerism in utero are unknown. The current study attempted to take advantage of this route for prenatal delivery of alpha-N-acetylglucosaminidase (Naglu) enzyme into the enzyme-deficient mouse model of Sanfilippo syndrome type B (MPS III B). Enzymatically sufficient mononuclear cells from human umbilical cord blood (MNC hUCB) were intravenously administered into heterozygote females modeling MPS III B on the 5th day of pregnancy during blastocyst implantation. The major findings were 1) administered MNC hUCB cells transmigrated and diffused into the embryos (E12.5); 2) some transmigrated cells expressed CD34 and CD117 antigens; 3) transmigrated cells were found in both the maternal and embryonic parts of placentas; 4) transmigrated cells corrected Naglu enzyme activity in all embryos; 5) administered MNC hUCB cells were extensively distributed in the organs and the blood of heterozygote mothers at one week after transplantation. Results indicate that prenatal delivery of Naglu enzyme by MNC hUCB cell administration into mothers of enzyme-deficient embryos is possible and may present a significant opportunity for new biotechnologies to treat many inherited disorders.


Subject(s)
Acetylglucosaminidase/genetics , Cord Blood Stem Cell Transplantation , Fetal Therapies , Leukocytes, Mononuclear/transplantation , Maternal-Fetal Exchange , Mucopolysaccharidosis III/therapy , Acetylglucosaminidase/deficiency , Animals , Antigens, CD34/analysis , Cell Lineage , Cell Movement , Female , Fetal Therapies/methods , Humans , Leukocytes, Mononuclear/enzymology , Male , Mice , Mice, Inbred C57BL , Models, Animal , Mucopolysaccharidosis III/embryology , Mucopolysaccharidosis III/enzymology , Mucopolysaccharidosis III/genetics , Placenta/ultrastructure , Pregnancy , Proto-Oncogene Proteins c-kit/analysis , Transplantation, Heterologous
19.
Ann N Y Acad Sci ; 1049: 67-83, 2005 May.
Article in English | MEDLINE | ID: mdl-15965108

ABSTRACT

Human umbilical cord blood (HUCB) is now considered a valuable source for stem cell-based therapies. HUCB cells are enriched for stem cells that have the potential to initiate and maintain tissue repair. This potential is especially attractive in neural diseases for which no current cure is available. Furthermore, HUCB cells are easily available and less immunogenic compared to other sources for stem cell therapy such as bone marrow. Accordingly, the number of cord blood transplants has doubled in the last year alone, especially in the pediatric population. The therapeutic potential of HUCB cells may be attributed to inherent ability of stem cell populations to replace damaged tissues. Alternatively, various cell types within the graft may promote neural repair by delivering neural protection and secretion of neurotrophic factors. In this review, we evaluate the preclinical studies in which HUCB was applied for treatment of neurodegenerative diseases and for traumatic and ischemic brain damage. We discuss how transplantation of HUCB cells affects these disorders and we present recent clinical studies with promising outcome.


Subject(s)
Brain Injuries/therapy , Cord Blood Stem Cell Transplantation , Fetal Blood/cytology , Neurodegenerative Diseases/therapy , Stem Cells/metabolism , Animals , Brain Injuries/pathology , Brain Ischemia/therapy , Cell Differentiation , Clinical Trials as Topic , Humans , Neurodegenerative Diseases/pathology , Stem Cells/cytology , Transplantation, Heterologous
20.
J Neurosurg Anesthesiol ; 17(2): 82-5, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15840993

ABSTRACT

Interleukin-1beta (IL-1beta) and interleukin-6 (IL-6) are major inflammatory cytokines produced after spinal cord injury (SCI). This study sought to evaluate the effects of methylprednisolone (MP) on IL-1beta and IL-6 protein in spinal cord tissue following SCI. Halothane-anesthetized, female Sprague-Dawley rats weighing (280-320 g) underwent laminectomy at T7-T8. No lesions were produced in animals in the saline control and MP control groups. SCI was induced by temporary placement of an aneurysm clip at T7-T8, with a closing pressure of 55 g at the spinal level of T7-T8, resulting in spinal cord compression for one minute. Animals with SCI were treated with MP (30 mg/kg sc) or an equal volume of saline. IL-1beta and IL-6 spinal cord protein were measured by enzyme-linked immunosorbent assays (ELISA). Data were summarized as mean +/- SD and compared by two-way analysis of variance (ANOVA). IL-1beta and IL-6 levels were elevated in the SCI + Saline animals (P < 0.01) compared with saline control, MP control, and SCI + MP-treated animals. The rise in IL-1beta and IL-6 levels after SCI was blunted after administration of MP, suggesting an interaction between glucocorticosteroids and the cytokine cascade after spinal cord trauma. Further evaluation of the effects of MP on the cytokine cascade may be important in assessing whether or not the anti-inflammatory effects of glucocorticosteroids confer neuroprotection after SCI.


Subject(s)
Interleukin-1/biosynthesis , Interleukin-6/biosynthesis , Methylprednisolone/pharmacology , Neuroprotective Agents/pharmacology , Spinal Cord Compression/metabolism , Spinal Cord/metabolism , Animals , Female , Nerve Tissue Proteins/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...