Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
J Exp Med ; 204(8): 1741-8, 2007 Aug 06.
Article in English | MEDLINE | ID: mdl-17635954

ABSTRACT

Upon cerebral hypoxia-ischemia (HI), apoptosis-inducing factor (AIF) can move from mitochondria to nuclei, participate in chromatinolysis, and contribute to the execution of cell death. Previous work (Cande, C., N. Vahsen, I. Kouranti, E. Schmitt, E. Daugas, C. Spahr, J. Luban, R.T. Kroemer, F. Giordanetto, C. Garrido, et al. 2004. Oncogene. 23:1514-1521) performed in vitro suggests that AIF must interact with cyclophilin A (CypA) to form a proapoptotic DNA degradation complex. We addressed the question as to whether elimination of CypA may afford neuroprotection in vivo. 9-d-old wild-type (WT), CypA(+/-), or CypA(-/-) mice were subjected to unilateral cerebral HI. The infarct volume after HI was reduced by 47% (P = 0.0089) in CypA(-/-) mice compared with their WT littermates. Importantly, CypA(-/-) neurons failed to manifest the HI-induced nuclear translocation of AIF that was observed in WT neurons. Conversely, CypA accumulated within the nuclei of damaged neurons after HI, and this nuclear translocation of CypA was suppressed in AIF-deficient harlequin mice. Immunoprecipitation of AIF revealed coprecipitation of CypA, but only in injured, ischemic tissue. Surface plasmon resonance revealed direct molecular interactions between recombinant AIF and CypA. These data indicate that the lethal translocation of AIF to the nucleus requires interaction with CypA, suggesting a model in which two proteins that normally reside in separate cytoplasmic compartments acquire novel properties when moving together to the nucleus.


Subject(s)
Active Transport, Cell Nucleus , Apoptosis Inducing Factor/metabolism , Cyclophilin A/physiology , Hypoxia-Ischemia, Brain/metabolism , Neurons/metabolism , Animals , Apoptosis , Brain/pathology , Cell Nucleus/metabolism , Cytoplasm/metabolism , Mice , Mice, Transgenic , Models, Biological , Oxidative Stress , Protein Binding
2.
Proc Natl Acad Sci U S A ; 105(38): 14632-7, 2008 Sep 23.
Article in English | MEDLINE | ID: mdl-18765809

ABSTRACT

Cranial radiation therapy is commonly used in the treatment of childhood cancers. It is associated with cognitive impairments tentatively linked to the hippocampus, a neurogenic region of the brain important in memory function and learning. Hippocampal neurogenesis is positively regulated by voluntary exercise, which is also known to improve hippocampal-dependent cognitive functions. In this work, we irradiated the brains of C57/BL6 mice on postnatal day 9 and evaluated both the acute effects of irradiation and the effects of voluntary running on hippocampal neurogenesis and behavior 3 months after irradiation. Voluntary running significantly restored precursor cell and neurogenesis levels after a clinically relevant, moderate dose of irradiation. We also found that irradiation perturbed the structural integration of immature neurons in the hippocampus and that this was reversed by voluntary exercise. Furthermore, irradiation-induced behavior alterations observed in the open-field test were ameliorated. Together, these results clearly demonstrate the usefulness of physical exercise for functional and structural recovery from radiation-induced injury to the juvenile brain, and they suggest that exercise should be evaluated in rehabilitation therapy of childhood cancer survivors.


Subject(s)
Cranial Irradiation , Hippocampus/cytology , Hippocampus/radiation effects , Neurons/cytology , Neurons/radiation effects , Running/physiology , Animals , Behavior, Animal/radiation effects , Cell Proliferation/radiation effects , Dentate Gyrus/cytology , Dentate Gyrus/physiology , Doublecortin Domain Proteins , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/genetics , Neurons/metabolism , Neuropeptides/genetics , Stem Cells/cytology
3.
J Neurochem ; 109(3): 858-66, 2009 May.
Article in English | MEDLINE | ID: mdl-19425175

ABSTRACT

Although the potential of adult neural stem cells to repair damage via cell replacement has been widely reported, the ability of endogenous stem cells to positively modulate damage is less well studied. We investigated whether medium conditioned by adult hippocampal stem/progenitor cells altered the extent of excitotoxic cell death in hippocampal slice cultures. Conditioned medium significantly reduced cell death following 24 h of exposure to 10 microM NMDA. Neuroprotection was greater in the dentate gyrus, a region neighboring the subgranular zone where stem/progenitor cells reside compared with pyramidal cells of the cornis ammonis. Using mass spectrometric analysis of the conditioned medium, we identified a pentameric peptide fragment that corresponded to residues 26-30 of the insulin B chain which we termed 'pentinin'. The peptide is a putative breakdown product of insulin, a constituent of the culture medium, and may be produced by insulin-degrading enzyme, an enzyme expressed by the stem/progenitor cells. In the presence of 100 pM of synthetic pentinin, the number of mature and immature neurons killed by NMDA-induced toxicity was significantly reduced in the dentate gyrus. These data suggest that progenitors in the subgranular zone may convert exogenous insulin into a peptide capable of protecting neighboring neurons from excitotoxic injury.


Subject(s)
Adult Stem Cells/drug effects , Excitatory Amino Acid Agonists/toxicity , N-Methylaspartate/toxicity , Neurons/drug effects , Neuroprotective Agents/pharmacology , Peptide Fragments/pharmacology , Animals , Animals, Newborn , Culture Media, Conditioned/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Hippocampus/cytology , Insulin/chemistry , Mass Spectrometry/methods , Organ Culture Techniques , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley
4.
Endocrinology ; 149(5): 2191-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18218693

ABSTRACT

Progenitor cells in the subgranular zone of the hippocampus may be of significance for functional recovery after various injuries because they have a regenerative potential to form new neuronal cells. The hippocampus has been shown to express the GH secretagogue (GHS) receptor 1a, and recent studies suggest GHS to both promote neurogenesis and have neuroprotective effects. The aim of the present study was to investigate whether GHS could stimulate cellular proliferation and exert cell protective effects in adult rat hippocampal progenitor (AHP) cells. Both hexarelin and ghrelin stimulated increased incorporation of (3)H-thymidine, indicating an increased cell proliferation. Furthermore, hexarelin, but not ghrelin, showed protection against growth factor deprivation-induced apoptosis, as measured by annexin V binding and caspase-3 activity and also against necrosis, as measured by lactate dehydrogenase release. Hexarelin activated the MAPK and the phosphatidylinositol 3-kinase/Akt pathways, whereas ghrelin activated only the MAPK pathway. AHP cells did not express the GHS receptor 1a, but binding studies could show specific binding of both hexarelin and ghrelin, suggesting effects to be mediated by an alternative GHS receptor subtype. In conclusion, our results suggest a differential effect of hexarelin and ghrelin in AHP cells. We have demonstrated stimulation of (3)H-thymidine incorporation with both hexarelin and ghrelin. Hexarelin, but not ghrelin, also showed a significant inhibition of apoptosis and necrosis. These results suggest a novel cell protective and proliferative role for GHS in the central nervous system.


Subject(s)
Cell Proliferation/drug effects , Cytoprotection/drug effects , Growth Hormone-Releasing Hormone/analogs & derivatives , Growth Hormone/metabolism , Hippocampus/drug effects , Stem Cells/drug effects , Animals , Cells, Cultured , Ghrelin/analogs & derivatives , Ghrelin/pharmacology , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Necrosis , Oligopeptides/pharmacology , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Rats , Receptors, Ghrelin/metabolism , Signal Transduction/drug effects , Stem Cells/pathology , Stem Cells/physiology
5.
Stem Cells ; 25(10): 2619-27, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17628017

ABSTRACT

After neurotrauma, ischemia, or neurodegenerative disease, astrocytes upregulate their expression of the intermediate filament proteins glial fibrillary acidic protein (GFAP), vimentin (Vim), and nestin. This response, reactive gliosis, is attenuated in GFAP(-/-)Vim(-/-) mice, resulting in the promotion of synaptic regeneration after neurotrauma and improved integration of retinal grafts. Here we assessed whether GFAP(-/-)Vim(-/-) astrocytes affect the differentiation of neural progenitor cells. In coculture with GFAP(-/-)Vim(-/-) astrocytes, neural progenitor cells increased neurogenesis by 65% and astrogenesis by 124%. At 35 days after transplantation of neural progenitor cells into the hippocampus, adult GFAP(-/-)Vim(-/-) mice had more transplant-derived neurons and astrocytes than wild-type controls, as well as increased branching of neurite-like processes on transplanted cells. Wnt3 immunoreactivity was readily detected in hippocampal astrocytes in wild-type but not in GFAP(-/-)Vim(-/-) mice. These findings suggest that GFAP(-/-)Vim(-/-) astrocytes allow more neural progenitor cell-derived neurons and astrocytes to survive weeks after transplantation. Thus, reactive gliosis may adversely affect the integration of transplanted neural progenitor cells in the brain. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Astrocytes/cytology , Glial Fibrillary Acidic Protein/deficiency , Hippocampus/cytology , Multipotent Stem Cells/transplantation , Neurons/cytology , Vimentin/deficiency , Animals , Astrocytes/physiology , Brain Tissue Transplantation , Cell Differentiation , Cells, Cultured/cytology , Coculture Techniques , Genes, RAG-1 , Glial Fibrillary Acidic Protein/genetics , Gliosis/genetics , Gliosis/pathology , Graft Survival , Mice , Mice, Inbred C57BL , Mice, Knockout , Multipotent Stem Cells/cytology , Oligodendroglia/cytology , Rats , Vimentin/genetics
6.
Endocrinology ; 148(8): 3765-72, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17510237

ABSTRACT

We have previously shown that recombinant human (rh) IGF-I induces cell proliferation and neurogenesis in the hippocampus of hypophysectomized rats. In the current investigation, we determined the effects of rhIGF-I on proliferation and differentiation in the cerebral cortex. Adult hypophysectomized rats were injected with bromodeoxyuridine (BrdU) to label newborn cells (once a day for the first 5 d), and rhIGF-I was administered peripherally for 6 or 20 d. In the cerebral cortex, the number of BrdU-labeled cells increased after 20 d but not after 6 d of rhIGF-I infusion. This suggests that rhIGF-I enhances the survival of newborn cells in the cerebral cortex. Using BrdU labeling combined with the oligodendrocyte-specific markers myelin basic protein and 2',3'-cyclic nucleotide 3'-phosphodiesterase, we demonstrated an increase in oligodendrogenesis in the cerebral cortex. The total amount of myelin basic protein and 2',3'-cyclic nucleotide 3'-phosphodiesterase was also increased on Western blots of homogenates of the cerebral cortex, confirming the immunohistochemical findings. Also, we observed an increase in the number of capillary-associated BrdU-positive cells, although total capillary area was not increased. rhIGF-I treatment did not affect cortical astrogliogenesis and neurogenesis was not observed. The ability of rhIGF-I to induce cortical oligodendrogenesis may have implications for the regenerative potential of the cortex.


Subject(s)
Cerebral Cortex/cytology , Hypophysectomy , Insulin-Like Growth Factor I/pharmacology , Oligodendroglia/cytology , Oligodendroglia/drug effects , Age Factors , Animals , Antimetabolites/pharmacokinetics , Astrocytes/cytology , Astrocytes/drug effects , Bromodeoxyuridine/pharmacokinetics , Capillaries , Cell Count , Cell Division/drug effects , Cell Survival/drug effects , Cerebral Cortex/blood supply , Female , Injections, Subcutaneous , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology
7.
J Cereb Blood Flow Metab ; 27(4): 785-94, 2007 Apr.
Article in English | MEDLINE | ID: mdl-16926844

ABSTRACT

The effects of hypoxia-ischemia (HI) on proliferation and differentiation in the immature (postnatal day 9) and juvenile (postnatal day 21) mouse hippocampus were investigated by injecting bromodeoxyuridine (50 mg/kg) daily for 7 days after the insult and evaluating the labeling 5 weeks after HI. Phenotypic differentiation was evaluated using NeuN, Iba1, APC, and S100beta as markers of neurons, microglia, oligodendrocytes, and astrocytes, respectively. The basal proliferation, in particular neurogenesis, was higher in the immature than in the juvenile hippocampus. Hypoxia-ischemia did not increase neurogenesis significantly in the immature dentate gyrus (DG), but it increased several-fold in the juvenile brain, reaching the same level as in the normal, noninjured immature brain. This suggests that the immature hippocampus is already working at the top of its proliferative capacity and that even though basal neurogenesis decreased with age, the injury-induced generation of new neurons in the juvenile hippocampus could not increase beyond the basal level of the immature brain. Generation of glial cells of all three types after HI was significantly more pronounced in the cornu ammonis of the hippocampus region of the juvenile hippocampus. In the DG, only microglia production was greater in the juvenile brain. Increased microglia proliferation correlated with increased levels of the proinflammatory cytokines MCP-1 and IL-18 3 days after HI, indicating that the inflammatory response is stronger in the juvenile hippocampus. In summary, contrary to what has been generally assumed, our results indicate that the juvenile brain has a greater capacity for neurogenesis after injury than the immature brain.


Subject(s)
Aging/pathology , Hypoxia-Ischemia, Brain/pathology , Inflammation/pathology , Neurons/pathology , Animals , Antimetabolites , Bromodeoxyuridine , Cell Count , Cell Differentiation/drug effects , Cell Proliferation , DNA-Binding Proteins , Dentate Gyrus/pathology , Hippocampus/pathology , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Neuroglia/pathology , Nuclear Proteins/genetics
8.
Eur J Neurosci ; 26(12): 3402-10, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18052985

ABSTRACT

We demonstrate that X chromosome-linked inhibitor of apoptosis protein (XIAP) counteracts oxidative stress in two essentially different disease-related models of brain injury, hypoxia-ischemia and irradiation, as judged by lower expression of nitrotyrosine (5-fold) and 4-hydroxy-2-nonenal (10-fold) in XIAP-overexpressing compared with wild-type mice. XIAP overexpression induced up-regulation of at least three antioxidants residing in mitochondria, superoxide dismutase 2, thioredoxin 2 and lysine oxoglutarate reductase. Cytochrome c release from mitochondria was reduced in XIAP-overexpressing mice. Hence, in addition to blocking caspases, XIAP can regulate reactive oxygen species in the brain, at least partly through up-regulation of mitochondrial antioxidants. XIAP-induced prevention of oxidative stress was not secondary to tissue protection because although XIAP overexpression provides tissue protection after hypoxia-ischemia, it does not prevent tissue loss after irradiation. This is a previously unknown role of XIAP and may provide the basis for development of novel protective strategies for both acute and chronic neurodegenerative diseases, where oxidative stress is an integral component of the injury mechanisms involved.


Subject(s)
Antioxidants/metabolism , Brain Ischemia/metabolism , Brain/radiation effects , Hypoxia, Brain/metabolism , Mitochondria/metabolism , Oxidative Stress , X-Linked Inhibitor of Apoptosis Protein/metabolism , Aldehydes/metabolism , Animals , Brain/metabolism , Cytochromes c/antagonists & inhibitors , Female , Gene Expression , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Up-Regulation
9.
J Neurosci ; 25(7): 1816-25, 2005 Feb 16.
Article in English | MEDLINE | ID: mdl-15716418

ABSTRACT

The hippocampal dentate gyrus (DG) is an area of active proliferation and neurogenesis within the adult brain. The molecular events controlling adult cell genesis in the hippocampus essentially remain unknown. It has been reported previously that adult male and female rats from the strains Sprague Dawley (SD) and spontaneously hypertensive (SHR) have a marked difference in proliferation rates of cells in the hippocampal DG. To exploit this natural variability and identify potential regulators of cell genesis in the hippocampus, hippocampal gene expression from male SHR as well as male and female SD rats was analyzed using a cDNA array strategy. Hippocampal expression of the gene-encoding glucose-dependent insulinotropic polypeptide (GIP) varied strongly in parallel with cell-proliferation rates in the adult rat DG. Moreover, robust GIP immunoreactivity could be detected in the DG. The GIP receptor is expressed by cultured adult hippocampal progenitors and throughout the granule cell layer of the DG, including progenitor cells. Thus, these cells have the ability to respond to GIP. Indeed, exogenously delivered GIP induced proliferation of adult-derived hippocampal progenitors in vivo as well as in vitro, and adult GIP receptor knock-out mice exhibit a significantly lower number of newborn cells in the hippocampal DG compared with wild-type mice. This investigation demonstrates the presence of GIP in the brain for the first time and provides evidence for a regulatory function for GIP in progenitor cell proliferation.


Subject(s)
Dentate Gyrus/metabolism , Gastric Inhibitory Polypeptide/physiology , Stem Cells/cytology , Animals , Cell Division/drug effects , Dentate Gyrus/cytology , Female , Gastric Inhibitory Polypeptide/biosynthesis , Gastric Inhibitory Polypeptide/genetics , Gastric Inhibitory Polypeptide/pharmacology , Gene Expression Profiling , Hypertension/genetics , Hypertension/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Neurons/drug effects , Oligonucleotide Array Sequence Analysis , Rats , Rats, Inbred SHR , Rats, Sprague-Dawley , Receptors, Gastrointestinal Hormone/deficiency , Receptors, Gastrointestinal Hormone/genetics , Receptors, Gastrointestinal Hormone/physiology
10.
Stroke ; 36(6): 1278-82, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15879324

ABSTRACT

BACKGROUND AND PURPOSE: The subventricular zone in the adult brain is identified as an endogenous resource of neuronal precursors that can be recruited to adjacent lesioned areas. The hypothesis was tested that postischemic environmental enrichment might enhance subventricular zone cell genesis. METHODS: A cortical infarct was induced in adult spontaneously hypertensive rats by ligating the middle cerebral artery distal to the striatal branches, after which animals were housed in either standard or enriched environment and allowed to survive for 5 weeks. The thymidine analogue bromodeoxyuridine was administered during the first postischemic week. The generation of neural stem/progenitor cells and neuronal precursors in the subventricular zone were studied with cell specific markers such as Ki67 and phosphorylated histone H3 (cell proliferation), Sox-2 (neural stem/progenitor cells), bromodeoxyuridine (slowly cycling, nonmigratory putative neural stem cells), and doublecortin (newborn immature neurons). RESULTS: Proliferating cells in the subventricular zone were identified as chiefly neural progenitors but also putative neural stem cells and neuronal precursors. Five weeks after stroke, proliferation in the subventricular zone was lower in stroke-lesioned rats housed in standard environment compared with nonlesioned rats. Postischemic environmental enrichment normalized cell proliferation levels, increased the numbers of putative neural stem cells as assessed with bromodeoxyuridine, and increased doublecortin-positive neuroblasts, which extended in migratory chains toward the infarct. CONCLUSIONS: Enriched environment increased the neural stem/progenitor cell pool and neurogenesis in the adult subventricular zone 5 weeks after a cortical stroke. This might be of potential importance for tissue regeneration.


Subject(s)
Brain Ischemia/pathology , Neurons/pathology , Stem Cells/metabolism , Stroke/pathology , Animals , Behavior, Animal , Biomarkers , Brain/metabolism , Brain/pathology , Brain Ischemia/metabolism , Bromodeoxyuridine/pharmacology , Cell Proliferation , DNA-Binding Proteins/metabolism , Doublecortin Domain Proteins , Doublecortin Protein , HMGB Proteins/metabolism , Histones/metabolism , Immunohistochemistry , Ki-67 Antigen/biosynthesis , Male , Microtubule-Associated Proteins/metabolism , Middle Cerebral Artery/pathology , Nervous System/pathology , Neurons/metabolism , Neuropeptides/metabolism , Phenotype , Rats , Rats, Inbred SHR , Regeneration , SOXB1 Transcription Factors , Stem Cells/cytology , Thymidine/chemistry , Time Factors , Transcription Factors/metabolism
11.
Neurosci Res ; 52(1): 1-9, 2005 May.
Article in English | MEDLINE | ID: mdl-15811547

ABSTRACT

It has previously been suggested that exogenous growth hormone (GH) affect quality of life and higher brain functions through the endogenous opioid system. Recently, we showed that GH down-regulate 72 and 48 kDa delta opioid receptor (DOR) proteins in the adult rat cerebral cortex and cerebellum. In the present study, we found that an antiserum raised against the N-terminus of the DOR also recognizes a 36 kDa protein, not recognized by a C-terminus-directed antiserum. We aimed to investigate the identity of the 72, 48 and 36 kDa proteins and to further study the effects of GH on their expression in different brain regions. The expression was studied in hypophysectomized (Hx) and untreated normal female rats. One subgroup of Hx rats received GH as a daily subcutaneous injection for 19 days. Our data show that treatment with GH in Hx rats normalized the expression of the 72 kDa protein in the cerebral cortex, whereas no significant effect were observed for the 48 or 36 kDa proteins. However, GH significantly reduced the ratio between the 72 and 36 kDa proteins in different brain regions of Hx rats. Our data suggest that GH reduces the levels of a 72 kDa DOR that likely represents a dimeric form of a 36 kDa DOR post-translationally truncated at the C-terminus, and that altered receptor dimerization may be involved in GH induced effects in the central nervous system.


Subject(s)
Brain/metabolism , Growth Hormone/pharmacology , Protein Isoforms/biosynthesis , Receptors, Opioid, delta/biosynthesis , Animals , Blotting, Western , Brain/drug effects , Female , Growth Hormone/metabolism , Hypophysectomy , Protein Isoforms/chemistry , Protein Isoforms/drug effects , Protein Processing, Post-Translational , Rats , Receptors, Opioid, delta/chemistry , Receptors, Opioid, delta/drug effects
12.
J Cereb Blood Flow Metab ; 22(7): 852-60, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12142570

ABSTRACT

The study aimed to elucidate the effects of cortical ischemia and postischemic environmental enrichment on hippocampal cell genesis. A cortical infarct was induced by a permanent ligation of the middle cerebral artery distal to the striatal branches in 6-month-old spontaneously hypertensive rats. Bromodeoxyuridine (BrdU) was administered as 7 consecutive daily injections starting 24 hours after surgery and animals were housed in standard or enriched environment. Four weeks after completed BrdU administration, BrdU incorporation and its co-localization with the neuronal markers NeuN and calbindin D28k, and the astrocytic marker glial fibrillary acidic protein in the granular cell layer and subgranular zone of the hippocampal dentate gyrus were determined with immunohistochemistry and were quantified stereologically. Compared with sham-operated rats, rats with cortical infarcts had a five-to sixfold ipsilateral increase in BrdU-labeled cells. About 80% of the new cells were neurons. Differential postischemic housing did not influence significantly the total number of surviving BrdU-labeled cells or newborn neurons. However, postischemic environmental enrichment increased the ipsilateral generation of astrocytes normalizing the astrocyte-to-neuron ratio, which was significantly reduced in rats housed in standard environment postischemically.


Subject(s)
Brain Ischemia/pathology , Cell Differentiation , Hippocampus/pathology , Animals , Animals, Newborn , Astrocytes/chemistry , Astrocytes/pathology , Biomarkers/analysis , Brain Infarction/pathology , Bromodeoxyuridine/analysis , Bromodeoxyuridine/metabolism , Calbindin 1 , Calbindins , Fluorescent Antibody Technique , Glial Fibrillary Acidic Protein/analysis , Immunohistochemistry , Male , Neurons/chemistry , Neurons/pathology , Phenotype , Rats , Rats, Inbred SHR , S100 Calcium Binding Protein G/analysis
13.
Brain Res Dev Brain Res ; 134(1-2): 115-22, 2002 Mar 31.
Article in English | MEDLINE | ID: mdl-11947942

ABSTRACT

In most brain regions of highly developed mammals, the majority of neurogenesis is terminated soon after birth. However, new neurons are continually generated throughout life in the subventricular zone and the dentate gyrus of the hippocampus. Insulin-like growth factor-I (IGF-I) is a polypeptide hormone that has demonstrated effects on these progenitor cells. IGF-I induces proliferation of isolated progenitors in culture, as well as affecting various aspects of neuronal induction and maturation. Moreover, systemic infusion of IGF-I increases both proliferation and neurogenesis in the adult rat hippocampus, and uptake of serum IGF-I by the brain parenchyma mediates the increase in neurogenesis induced by exercise. Neurogenesis in the adult brain is regulated by many factors including aging, chronic stress, depression and brain injury. Aging is associated with reductions in both hippocampal neurogenesis and IGF-I levels, and administration of IGF-I to old rats increases neurogenesis and reverses cognitive impairments. Similarly, stress and depression also inhibit neurogenesis, possibly via the associated reductions in serotonin or increases in circulating glucocorticoids. As both of these changes have the potential to down regulate IGF-I production by neural cells, stress may inhibit neurogenesis indirectly via downregulation of IGF-I. In contrast, brain injury stimulates neurogenesis, and is associated with upregulation of IGF-I in the brain. Thus, there is a tight correlation between IGF-I and neurogenesis in the adult brain under different conditions. Further studies are needed to clarify whether IGF-I does indeed mediate neurogenesis in these situations.


Subject(s)
Aging/physiology , Brain/cytology , Insulin-Like Growth Factor I/physiology , Neurons/cytology , Animals , Cell Division/physiology
14.
Neurosci Lett ; 369(1): 24-7, 2004 Oct 07.
Article in English | MEDLINE | ID: mdl-15380301

ABSTRACT

Sox-2 is a transcription factor that is expressed by self-renewing and multipotent stem cells of the embryonic neuroepithelium. Very little is however known about Sox-2 expression in the adult brain and therefore we used immunohistochemistry to examine its distribution and co-localization with specific cell markers. We found that Sox-2 was expressed by actively dividing neural progenitor cells in the neurogenic regions in the adult rat brain, the subventricular zone of the forebrain and the subgranular zone of the dentate gyrus in the hippocampus. Cells expressing immature neuronal markers were essentially Sox-2 immunonegative. Sox-2 was also found to be expressed by glial fibrillary acidic protein immunopositive astroglia, widely distributed in the brain parenchyma. Given the fact that several studies have established the neurogenic capacity of a specialized type of astroglia in the adult brain, the findings of Sox-2 expression in parenchymal astroglia are of potential interest. We conclude that Sox-2 might, in combination with appropriate cell-specific markers, constitute a useful marker to study the in vivo dynamics of the neural progenitor cell compartment also in the adult brain.


Subject(s)
Astrocytes/metabolism , Brain/cytology , DNA-Binding Proteins/metabolism , Neurons/metabolism , Nuclear Proteins/metabolism , Stem Cells/metabolism , Animals , Brain/metabolism , Glial Fibrillary Acidic Protein/metabolism , HMGB Proteins , Immunohistochemistry/methods , Ki-67 Antigen/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Rats , Rats, Sprague-Dawley , SOXB1 Transcription Factors , Sialic Acids/metabolism , Transcription Factors
15.
Neurosci Lett ; 354(2): 163-5, 2004 Jan 09.
Article in English | MEDLINE | ID: mdl-14698463

ABSTRACT

Oxidative stress plays an important role in the development of tissue damage following transient focal cerebral ischaemia. Glutathione is a central component in the antioxidant defence of cells. We have previously shown a close association between mitochondrial glutathione loss and cell death following middle cerebral artery (MCA) occlusion. Glutathione monoethyl ester increases cellular glutathione and is particularly effective in increasing the mitochondrial pool. In the present investigation, we infused glutathione monoethyl ester into the third ventricle during 2 h of MCA occlusion and 48 h of reperfusion. Infarct size was reduced from 46% of the total ischaemic hemisphere in saline-treated animals to 16% following ester treatment. Thus, glutathione monoethyl ester provides neuroprotection following transient focal cerebral ischaemia.


Subject(s)
Cerebral Infarction/prevention & control , Glutathione/analogs & derivatives , Glutathione/pharmacology , Ischemic Attack, Transient/drug therapy , Nerve Degeneration/prevention & control , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Animals , Brain/drug effects , Brain/pathology , Brain/physiopathology , Cerebral Infarction/physiopathology , Disease Models, Animal , Glutathione/metabolism , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/physiopathology , Intracellular Fluid/drug effects , Intracellular Fluid/metabolism , Ischemic Attack, Transient/metabolism , Ischemic Attack, Transient/physiopathology , Male , Mitochondria/drug effects , Mitochondria/metabolism , Nerve Degeneration/physiopathology , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , Treatment Outcome
16.
J Rehabil Med ; (41 Suppl): 17-9, 2003 May.
Article in English | MEDLINE | ID: mdl-12817652

ABSTRACT

Recent findings concerning the regenerative potential of the adult brain suggest a more pronounced plasticity than previously thought. One such finding is the generation of new neurons in the adult brain (neurogenesis). Loss of neurons has long been considered to be irreversible in the adult human brain, i.e., dead neurons are not replaced. The inability to generate replacement cells is thought to be an important cause of neurological disease and impairment. In most brain regions, the generation of neurons is generally confined to a discrete developmental period. Exceptions have recently been described in several regions of the brain that have been shown to generate new neurons well into the postnatal and adult period. One of the best characterized regions is the subgranular zone of the dentate gyrus in the brain, where granule neurons are generated throughout life from a population of progenitor/ stem cells. Furthermore, recent findings suggest that neurogenesis may be of importance for memory function as well as mood disorders. Several very important questions can be formulated on the basis of these discoveries, for instance, what factors influence the generation of new neurons and whether it is possible for enhanced neurogenesis to contribute to functional recovery.


Subject(s)
Dentate Gyrus/physiology , Nerve Regeneration/physiology , Neuronal Plasticity/physiology , Adult , Animals , Cell Division , Dentate Gyrus/cytology , Dentate Gyrus/growth & development , Environment , Humans , Infant, Newborn , Rats , Stem Cell Transplantation , Stem Cells/physiology
17.
Stem Cell Res ; 3(1): 39-50, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19515621

ABSTRACT

Tissue engineering and cell therapy require large-scale production of homogeneous populations of lineage-restricted progenitor cells that easily can be induced to differentiate into a specific tissue. We have developed straightforward protocols for the establishment of human embryonic stem (hES) cell-derived mesenchymal progenitor (hES-MP) cell lines. The reproducibility was proven by derivation of multiple hES-MP cell lines from 10 different hES cell lines. To illustrate clinical applicability, a xeno-free hES-MP cell line was also derived. None of the markers characteristic for undifferentiated hES cells were detected in the hES-MP cells. Instead, these cells were highly similar to mesenchymal stem cells with regard to morphology and expression of markers. The safety of hES-MP cells following transplantation was studied in severely combined immunodeficient (SCID) mice. The implanted hES-MP cells gave rise to homogeneous, well-differentiated tissues exclusively of mesenchymal origin and no teratoma formation was observed. These cells further have the potential to differentiate toward the osteogenic, adipogenic, and chondrogenic lineages in vitro. The possibility of easily and reproducibly generating highly expandable hES-MP cell lines from well-characterized hES cell lines with differentiation potential into several mesodermal tissues entails an enormous potential for the field of regenerative medicine.


Subject(s)
Embryonic Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Adipogenesis , Animals , Cell Differentiation , Cell Line , Cell Lineage , Chondrogenesis , Humans , Mesenchymal Stem Cell Transplantation , Mice , Mice, SCID , Osteogenesis , Regenerative Medicine , Teratoma/pathology
18.
Nat Rev Neurosci ; 8(9): 712-23, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17704813

ABSTRACT

During brain development, one of the most important structures is the subventricular zone (SVZ), from which most neurons are generated. In adulthood the SVZ maintains a pool of progenitor cells that continuously replace neurons in the olfactory bulb. Neurodegenerative diseases induce a substantial upregulation or downregulation of SVZ progenitor cell proliferation, depending on the type of disorder. Far from being a dormant layer, the SVZ responds to neurodegenerative disease in a way that makes it a potential target for therapeutic intervention.


Subject(s)
Lateral Ventricles/pathology , Lateral Ventricles/physiopathology , Neurodegenerative Diseases/pathology , Neurons/physiology , Animals , Humans , Neurodegenerative Diseases/physiopathology , Neurons/pathology
19.
Curr Pharm Biotechnol ; 8(3): 117-25, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17584084

ABSTRACT

Since the discovery of endogenous progenitor cells in two brain regions in the adult, the notion that progenitor cells might be useful for repairing damaged neurons or replacing dead neurons has gone from fiction to a reality, at least in the laboratory setting. Progenitor cells have the unique ability to be able to produce new neurons in response to endogenous and exogenous cues from their microenvironment in the brain and from the environment of the organism. However, in models of several disorders and insults the regenerative potential of the central nervous system need external enhancing. In this review we begin by focussing on the developments in the field of neurobiology that have led to the specific study of neural progenitor cell biology. In particular we discuss the two germinal niches, the subventricular zone and the subgranular zone, as well as how various neurological diseases affect these niches. We furthermore try to define primary progenitor cell disorders and secondary progenitor cell responses. The second part of this review focuses on proteomic approaches for studying progenitor cells. These techniques allow the array of proteins that are expressed by progenitor cells to be determined and further more allow comparisons between diseased and normal cells or treated and untreated cell populations. If we can induce neural progenitor cells to generate functional neurons in the central nervous system (CNS) then the burden of neurological disorders may be eased in the future. The advances in proteomic technology have and will enable further understanding of the regulatory processes in these cells so that progenitor cell integration and differentiation can be enhanced. Hopefully an increase in knowledge of progenitor cell biology will have a major impact on clinical practice.


Subject(s)
Brain/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Proteome/metabolism , Stem Cells/metabolism , Stem Cells/pathology , Animals , Biomarkers/metabolism , Brain Diseases/metabolism , Brain Diseases/pathology , Humans
20.
Clin Exp Pharmacol Physiol ; 34(5-6): 528-32, 2007.
Article in English | MEDLINE | ID: mdl-17439428

ABSTRACT

1. The subventricular zone (SVZ) of the forebrain that overlies the caudate nucleus is one of the principal brain regions in which neurogenesis occurs in the human brain, throughout life. 2. In response to the degeneration that occurs in the caudate nucleus in Huntington's disease, or in the caudate nucleus or cortex in stroke models, the SVZ increases the production of progenitor cells that migrate towards the site of the damage where they can differentiate into mature neurons and glial cells. The SVZ contains three main cell types and these are progenitor cells, glial cells and migratory neuroblasts; glial cells are the most common cell type and, in response to Huntington's disease, most of the SVZ cell proliferation is glial, but the number of precursor and neuroblasts is also increased. 3. The SVZ is enriched in neuroactive compounds, such as neuropeptide Y and gamma-aminobutyric acid receptor subunits gamma2, which stimulate ongoing neurogenesis. Interestingly, these stimulating cues are upregulated in the SVZ in response to Huntington's disease. Thus, the SVZ comprises heterogeneous cell types that are maintained in an environment that is permissive to neurogenesis and gliogenesis, and responds to neurodegenerative changes in adjacent brain regions by increasing progenitor cell proliferation and neurogenesis in an attempt to replace the cells that die as a result of neurodegeneration.


Subject(s)
Basal Ganglia/physiopathology , Neurodegenerative Diseases/physiopathology , Neurons/physiology , Stem Cells/physiology , Adult , Animals , Basal Ganglia/injuries , Brain/pathology , Brain/physiopathology , Disease Models, Animal , Humans , Models, Neurological , Neurodegenerative Diseases/pathology , Neurons/cytology , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL