Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Int J Clin Exp Pathol ; 7(10): 6486-92, 2014.
Article in English | MEDLINE | ID: mdl-25400726

ABSTRACT

DSTYK (Dual serine/threonine and tyrosine protein kinase) is a putative dual Ser/Thr and Tyr protein kinase with unique structural features. It is proposed that DSTYK may play important roles in brain because of its high expression in most brain areas. In the present study, a DSTYK knockout (KO) mouse line with the ablation of C-terminal of DSTYK including the kinase domain was generated to study the physiological function of DSTYK. The DSTYK KO mice are fertile and have no significant morphological defects revealed by Nissl staining compared with wildtype mice. Open field test and rotarod test showed there is no obvious difference in basic motor and balance capacity between the DSTYK homozygous KO mice and DSTYK heterozygous KO mice. In water maze test, however, the DSTYK homozygous KO mice show impaired capabilities of learning and memory compared with the DSTYK heterozygous KO mice.


Subject(s)
Behavior, Animal , Learning Disabilities/enzymology , Maze Learning , Memory Disorders/enzymology , Memory , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Animals , Genotype , Learning Disabilities/genetics , Learning Disabilities/psychology , Male , Memory Disorders/genetics , Memory Disorders/psychology , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Phenotype , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Rotarod Performance Test
2.
Int J Clin Exp Pathol ; 7(5): 2179-84, 2014.
Article in English | MEDLINE | ID: mdl-24966926

ABSTRACT

Globin family was famous for oxygen supply function of its members such as hemoglobin and myoglobin. With the progress of research, several members of this protein family have been proven to play roles in tumors including glioma. Androglobin (ADGB) is a recently identified member of globin family with very few studies about its function. In the present study, we show that ADGB plays an oncogene role in glioma. Lentiviral vector mediated ADGB knockdown inhibited the proliferation of glioma cell lines determined by MTT assay and colony formation assay. ADGB knockdown also increased the apoptosis of glioma cell line U251 assessed by flow cytometry. In addition, western blot showed that ADGB knockdown altered levels of several proteins related to proliferation, survival or apoptosis in U251 cells. These findings suggest ADGB is involved in the progression of glioma in vitro.


Subject(s)
Brain Neoplasms/metabolism , Calmodulin-Binding Proteins/metabolism , Cell Proliferation , Glioma/metabolism , Globins/metabolism , Apoptosis , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Calmodulin-Binding Proteins/genetics , Cell Line, Tumor , Down-Regulation , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Genetic Vectors , Glioma/genetics , Glioma/pathology , Globins/genetics , Humans , Lentivirus/genetics , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Time Factors , Transfection
3.
Brain Res Bull ; 100: 55-60, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24270001

ABSTRACT

Arcuate nucleus of hypothalamus (ARH) is the core component in the regulation circuits of food intake and energy homeostasis. ARH projections to other parts of the hypothalamus and to extrahypothalamic areas are established in the postnatal two weeks, which is a pivotal stage for individual development. ß-Catenin, a cell adhesion protein and also the mediator of canonical Wnt signaling pathway, plays an important role in embryonic development and adult homeostasis. However, whether ß-catenin plays any roles in the development of hypothalamus is not clear. Here, we report that perinatal conditional knockout of ß-catenin by CamKIIα-Cre in forebrain reduces body weight gain from P8 and dramatically shortens life span. Quantitative PCR and in situ hybridization results showed the expression of NPY mRNA in the ARH of ß-catenin CKO mice at P15 is obviously increased compared with that of littermate controls, whereas the expression of POMC mRNA is significantly decreased, which suggested the reduction of postnatal body weight gain might be due to the deficiency of food intake. Together, ß-catenin might play an important role in the regulation of food intake and postnatal body weight gain probably through affecting the development of ARH circuits.


Subject(s)
Arcuate Nucleus of Hypothalamus/physiology , Body Weight/genetics , Eating/genetics , Weight Gain/genetics , beta Catenin/genetics , Animals , In Situ Hybridization , Mice , Mice, Transgenic , Neuropeptide Y/metabolism , Pro-Opiomelanocortin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , beta Catenin/metabolism
4.
Int J Clin Exp Pathol ; 6(11): 2460-7, 2013.
Article in English | MEDLINE | ID: mdl-24228108

ABSTRACT

ZFX (zinc finger transcription factor, X chromosome-linked) contributes to the maintenance of different types of stem cells and the progression of various cancers. We have previously reported that ZFX knockdown inhibits proliferation of glioma in vitro and in vivo. Since overexpression of ZFX in lung cancer tissue correlates with lymph node metastasis, we hypothesized that ZFX may play a role in lung cancer. In this study, we identified ZFX as a promoter of lung cancer growth and migration in a NSCLC (non-small cell lung carcinoma) cell line H1299. ZFX knockdown caused proliferation inhibition determined by MTT assay and colony formation assay, G0/G1 arrest of cell cycle and slightly increased proportion of apoptotic cells assessed by flow cytometry assay, decreased population of migrating cells showed by wound-healing assay, increased cell senescence evidenced by senescence-associated ß-galactosidase staining. ZFX knockdown also led to decreased proportion of tumor bearing mice and reduced mean tumor volume in a subcutaneous tumor model. In addition, western blot showed that ZFX knockdown down regulated a set of proteins involved in proliferation, survival and motility. Altogether, these results suggest that ZFX may be a potential therapeutic target for NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/therapy , Cell Movement , Cell Proliferation , Genetic Therapy , Kruppel-Like Transcription Factors/metabolism , Lung Neoplasms/therapy , Animals , Apoptosis , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle Checkpoints , Cell Line, Tumor , Cellular Senescence , Gene Knockdown Techniques , Humans , Kruppel-Like Transcription Factors/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , RNA Interference , Time Factors , Transfection , Tumor Burden , Xenograft Model Antitumor Assays
5.
Int J Clin Exp Pathol ; 6(7): 1261-71, 2013.
Article in English | MEDLINE | ID: mdl-23826407

ABSTRACT

Somatosensory ganglia including dorsal root ganglion (DRG) and trigeminal ganglion (TG) are derived from a common pool of neural crest stem cells (NCCs), and are good systems to study the mechanisms of neurogenesis and gliogenesis. Previous studies have reported that deletion of Rbpj, a critical integrator of activation signals from all Notch receptors, in NCCs and their derived cells resulted in the delayed gliogenesis at early stage and a loss of glial cells at later stage in the DRG. But the phenotypes in the TG have not been described. Here we reported although the gliogenesis was also delayed initially in Rbpj-deficient TG, it was recovered as the development progressed, as shown by the presence of large number of glial cells in the TG at later stages. However, neuronal reduction was observed in Rbpj-deficient TG, which is similar to what observed in Rbpj-deficient DRG. Taken together, our data indicate the function of Rbpj is diversified and context dependent in the gliogenesis of somatosensory ganglia.


Subject(s)
Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Neuroglia/metabolism , Trigeminal Ganglion/metabolism , Animals , Biomarkers/metabolism , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Neurogenesis , Neurons/metabolism , Phenotype , Receptors, Nerve Growth Factor/metabolism , SOXE Transcription Factors/metabolism , Time Factors , Trigeminal Ganglion/embryology
6.
J Cell Sci ; 125(Pt 18): 4320-32, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22718343

ABSTRACT

The locus coeruleus (LC) is the main source of noradrenaline in the brain and is implicated in a broad spectrum of physiological and behavioral processes. However, genetic pathways controlling the development of noradrenergic neurons in the mammalian brain are largely unknown. We report here that Rbpj, a key nuclear effector in the Notch signaling pathway, plays an essential role in LC neuron development in the mouse. Conditional inactivation of Rbpj in the dorsal rhombomere (r) 1, where LC neurons are born, resulted in a dramatic increase in the number of Phox2a- and Phox2b-expressing early-differentiating LC neurons, and dopamine-ß-hydroxylase- and tyrosine-hydroxylase-expressing late-differentiating LC neurons. In contrast, other neuronal populations derived from the dorsal r1 were either reduced or unchanged. In addition, a drastic upregulation of Ascl1, an essential factor for noradrenergic neurogenesis, was observed in dorsal r1 of conditional knockout mice. Through genomic sequence analysis and EMSA and ChIP assays, a conserved Rbpj-binding motif was identified within the Ascl1 promoter. A luciferase reporter assay revealed that Rbpj per se could induce Ascl1 transactivation but this effect was counteracted by its downstream-targeted gene Hes1. Moreover, our in vitro gene transfection and in ovo electroporation assays showed that Rbpj upregulated Ascl1 expression when Hes1 expression was knocked down, although it also exerted a repressive effect on Ascl1 expression in the presence of Hes1. Thus, our results provide the first evidence that Rbpj functions as a key modulator of LC neuron development by regulating Ascl1 expression directly, and indirectly through its target gene Hes1.


Subject(s)
Adrenergic Neurons/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Locus Coeruleus/metabolism , Locus Coeruleus/pathology , Neurogenesis , Receptors, Notch/metabolism , Signal Transduction , Adrenergic Neurons/pathology , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Morphogenetic Proteins/metabolism , COUP Transcription Factors/metabolism , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Down-Regulation/genetics , Embryo, Mammalian/metabolism , Gene Deletion , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hyperplasia , Locus Coeruleus/embryology , Mice , Mice, Knockout , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Protein Binding , Repressor Proteins , Serotonergic Neurons/metabolism , Serotonergic Neurons/pathology , Stem Cells/metabolism
7.
Genesis ; 50(7): 552-60, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22368151

ABSTRACT

Prrxl1-CreER(T2) transgenic mice expressing tamoxifen-inducible Cre recombinase were generated by modifying a Prrxl1-containing BAC clone. Cre recombination activity was examined in Prrxl1-CreER(T2); Rosa26 reporter mice at various embryonic and postnatal stages. Pregnant mice were treated with a single dose of tamoxifen at embryonic day (E) 9.5 or E12.5, and X-gal staining was performed 2 days later. Strong X-gal staining was observed in the somatosensory ganglia (e.g., dorsal root and trigeminal ganglia) and the first central sites for processing somatosensory information (e.g., spinal dorsal horn and trigeminal nerve-associated nuclei). When tamoxifen was administered at postnatal day (P) 20 or in adulthood (P120), strong Cre recombination activity was present in the primary somatosensory ganglia, while weak Cre recombination activity was found in the spinal dorsal horn, mesencephalic trigeminal nucleus, principal sensory trigeminal nucleus, and spinal trigeminal nucleus. This mouse line provides a useful tool for exploring genes' functions in the somatosensory system in a time-controlled way.


Subject(s)
Afferent Pathways/physiology , Homeodomain Proteins/genetics , Mice, Transgenic , Nerve Tissue Proteins/genetics , Somatosensory Cortex/physiology , Spinal Nerve Roots/physiology , Transcription Factors/genetics , Trigeminal Ganglion/physiology , Afferent Pathways/embryology , Animals , Chromosomes, Artificial, Bacterial , Embryo, Mammalian , Female , Founder Effect , Gene Expression Regulation, Developmental/drug effects , Genes, Reporter , Integrases/genetics , Mice , Pregnancy , Promoter Regions, Genetic , Proteins/genetics , RNA, Untranslated , Recombination, Genetic/drug effects , Somatosensory Cortex/embryology , Spinal Nerve Roots/embryology , Tamoxifen/administration & dosage , Time Factors , Trigeminal Ganglion/embryology
8.
Neurosci Res ; 71(1): 12-21, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21658419

ABSTRACT

This study consists of a thorough immunohistochemical examination of the expression profile of the transcription factor Satb1 (special AT-rich sequence binding protein 1) in the adult mouse central nervous system (CNS). Satb1-positive neurons were abundant in the deep layers of the neocortex, subiculum, anterior olfactory nucleus, nucleus of diagonal band, anterior part of the basolateral amygdaloid nucleus, compact part of substantia nigra, ventral tegmental area, ventral and dorsal tegmental nuclei, laterodorsal tegmental nucleus, and medullary and spinal dorsal horns. Relatively smaller populations of Satb1-positive neurons were observed in the piriform cortex, hippocampus, other subnuclei of the amygdala, centrolateral thalamic nucleus, parafascicular thalamic nucleus, posterior hypothalamic area, ventral part of the premammillary nucleus, supramammillary nucleus, deep layers of the superior colliculus, dorsal raphe nucleus, nucleus of trapezoid body, superior periolivary nucleus and nucleus of lateral lemniscus, and parabrachial region. Double immunostaining showed that Satb1 was expressed in midbrain dopaminergic neurons, but not in cholinergic or serotonergic neurons. Satb1 expression was never observed in glial cells. This study presents a comprehensive overview of Satb1 expression in the CNS, and provides insights for investigating the role of Satb1 in the mature CNS.


Subject(s)
Central Nervous System/metabolism , Matrix Attachment Region Binding Proteins/metabolism , Neurons/metabolism , Animals , Biomarkers/metabolism , Central Nervous System/cytology , Choline O-Acetyltransferase/biosynthesis , Cholinergic Neurons/cytology , Cholinergic Neurons/metabolism , DNA-Binding Proteins , Dopaminergic Neurons/cytology , Dopaminergic Neurons/enzymology , Dopaminergic Neurons/metabolism , Female , Male , Matrix Attachment Region Binding Proteins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neuroglia/metabolism , Nuclear Proteins/metabolism , Serotonin/physiology
9.
Neural Dev ; 6: 14, 2011 Apr 21.
Article in English | MEDLINE | ID: mdl-21510873

ABSTRACT

BACKGROUND: The dorsal root ganglion (DRG) is composed of well-characterized populations of sensory neurons and glia derived from a common pool of neural crest stem cells (NCCs), and is a good system to study the mechanisms of neurogenesis and gliogenesis. Notch signaling is known to play important roles in DRG development, but the full scope of Notch functions in mammalian DRG development remains poorly understood. RESULTS: In the present study, we used Wnt1-Cre to conditionally inactivate the transcription factor Rbpj, a critical integrator of activation signals from all Notch receptors, in NCCs and their derived cells. Deletion of Rbpj caused the up-regulation of NeuroD1 and precocious neurogenesis in DRG early development but led to an eventual deficit of sensory neurons at later stages, due to reduced cell proliferation and abnormal cell death. In addition, gliogenesis was delayed initially, but a near-complete loss of glia was observed finally in Rbpj-deficient DRG. Furthermore, we found P75 and Sox10, which are normally expressed exclusively in neuronal and glial progenitors of the DRG after the NCCs have completed their migration, were co-expressed in many cells of the DRG of Rbpj conditional knock-out mice. CONCLUSIONS: Our data indicate that Rbpj-mediated canonical Notch signaling inhibits DRG neuronal differentiation, possibly by regulating NeuroD1 expression, and is required for DRG gliogenesis in vivo.


Subject(s)
Ganglia, Spinal , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Sensory Receptor Cells/physiology , Animals , Animals, Genetically Modified , Cell Death/genetics , Cell Movement/genetics , Cell Proliferation , Embryo, Mammalian , Ganglia, Spinal/cytology , Ganglia, Spinal/embryology , Ganglia, Spinal/growth & development , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/physiology , Receptor, trkC/metabolism , Up-Regulation/genetics , Wnt1 Protein/genetics
10.
Dev Biol ; 348(1): 87-96, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20875817

ABSTRACT

Netrin-1 and DCC are well known for their roles in neurite growth, axonal guidance, and neuronal migration. Recently, a number of studies showed that DCC is involved in the induction of apoptosis, and this proapoptotic activity can be blocked in the presence of Netrin-1. However, here, we found that DCC is required for the survival of two types of neurons selectively in the developing mouse retina where DCC is abundantly expressed. Our results showed that the DCC(-/-) retina displayed a reduced ganglion cell layer with relatively normal neuroblastic layer. Immunostaining assays revealed that in DCC(-/-) mice, initial neurogenesis within retina was unchanged while the numbers of differentiated retinal ganglion cells and displaced amacrine cells in ganglion cell layer were greatly reduced due to increased apoptosis. By contrast, other neuronal types including horizontal cells, bipolar cells, amacrine cells, photoreceptors, and Müller cells appeared normal in DCC mutant retinas. Moreover, DCC(kanga) mice that lack the intracellular P3 domain of DCC receptor displayed the same defects as DCC(-/-) mice. Thus, our findings suggest that DCC is a key regulator for the survival of specific types of neurons during retinal development and that DCC-P3 domain is essential for this developing event.


Subject(s)
Amacrine Cells/cytology , Eye Proteins/physiology , Neurogenesis/physiology , Receptors, Cell Surface/physiology , Retina/growth & development , Retinal Ganglion Cells/cytology , Tumor Suppressor Proteins/physiology , Amacrine Cells/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Survival , DCC Receptor , Eye Abnormalities/genetics , Eye Abnormalities/pathology , Eye Proteins/chemistry , Eye Proteins/genetics , Gene Expression Regulation, Developmental , Genes, DCC , Mice , Mice, Knockout , Mice, Mutant Strains , Neurogenesis/genetics , Protein Structure, Tertiary , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Retina/cytology , Retina/metabolism , Retinal Ganglion Cells/metabolism , Sequence Deletion , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
11.
Brain Res Bull ; 82(1-2): 74-82, 2010 Apr 29.
Article in English | MEDLINE | ID: mdl-20206242

ABSTRACT

Rhombomere 1 (r1), a temporary structure in the early developing brain, is bounded rostrally by the isthmus organizer and caudally by the r2 domain. Many genes involved in r1 induction and patterning have been identified. However, our knowledge of a comprehensive map of r1 regionalization defined by these genes at early embryonic stages remains fragmentary. In the present study, we examined the expression of a variety of genes in the dorsal r1 of E9.0-E10.5 mouse embryos. The expression domains within ventricular zone of these genes examined allowed us to define four distinct regions along the anterior-posterior (A-P) axis of the dorsal r1: the Wnt1/Lmx1a/Gdf7/Msx1/Msx2-positive roof plate, Math1/Olig3/Msx1/Msx2-positive, upper rhombic lip, Mash1/Ngn1/Ngn2-positive intermediate domain, and Mash1/Ngn1/Ngn2-positive rostral domain. Moreover, we defined the distribution of several genes expressed in the mantle zone of the dorsal r1, including Lmx1b, Lhx2, Lhx9, Phox2a and Phox2b. Taken together, our gene expression data identify a refined subdivision of the dorsal r1 with four distinct domains along the A-P axis and a mantle zone at early embryonic stages.


Subject(s)
Brain/embryology , Embryo, Mammalian , Gene Expression Regulation, Developmental , Morphogenesis , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/anatomy & histology , Brain/physiology , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , In Situ Hybridization , LIM-Homeodomain Proteins , MSX1 Transcription Factor/genetics , MSX1 Transcription Factor/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Wnt1 Protein/genetics , Wnt1 Protein/metabolism
12.
Brain Res ; 1289: 96-105, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19615349

ABSTRACT

Erythropoietin (Epo) has been gaining great interest for its potential neuroprotective effect in various neurological insults. However, the molecular mechanism underlying how Epo exerts the function is not clear. Recent studies have indicated that Zn(2+) may have a key role in selective cell death in excitotoxicity after injury. In the present study, we studied the effect of recombinant human Epo (rhEpo) in zinc-induced neurotoxicity both in vitro and in vivo. Exposure of cultured hippocampal neurons to 200 muM ZnC1(2) for 20 min resulted in remarkable neuronal injury, revealed by assessing neuronal morphology. By measuring mitochondrial function using MTT assay, we found that application of rhEpo (0.1 U/ml) 24 h before zinc exposure resulted in a significant increase of neuronal survival (0.6007+/-0.2280 Epo group vs 0.2333+/-0.1249 in control group; n=4, p<0.01). Furthermore, we demonstrated that administration of rhEpo (5,000 IU/kg, intraperitoneal) 30 min after traumatic brain injury (TBI) in rats dramatically protected neuronal death indicated by ZP4 staining, a new zinc-specific fluorescent sensor which has been widely used to indicate neuronal damage after excitotoxic injury (n=5/group, p<0.05). Neuronal damage was also assessed by Fluoro-Jade B (FJB) staining, a highly specific fluorescent marker for the degenerating neurons. Consistent with ZP4 staining, we found the beneficial effects of rhEpo on neuronal survival in hippocampus after TBI (n=5/group, p<0.05). Our results suggest that rhEpo can significantly reduce the pathological Zn(2+) accumulation in rat hippocampus after TBI as well as zinc-induced cell death in cultured cells, which may potentially contribute to its neuronal protection after excitotoxic brain damage.


Subject(s)
Brain Injuries/drug therapy , Brain Injuries/pathology , Cell Death/drug effects , Erythropoietin/therapeutic use , Hippocampus/pathology , Analysis of Variance , Animals , Brain Injuries/metabolism , Cell Count , Cells, Cultured , Chlorides/toxicity , Dose-Response Relationship, Drug , Erythropoietin/pharmacology , Fluoresceins/metabolism , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Immunohistochemistry , Male , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins , Zinc/metabolism , Zinc Compounds/toxicity
13.
Neurosci Res ; 61(4): 420-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18554735

ABSTRACT

GATA binding protein 3 (GATA3) is an important regulator of central nervous system (CNS) development, but its expression pattern in the postnatal CNS has not been studied. In the present study, we examined the distribution of GATA3 mRNA in the mouse CNS at different postnatal stages by in situ hybridization. During the first 2 weeks of postnatal development, numerous GATA3-expressing cells were found in the intergeniculate leaf, ventral lateral geniculate nucleus, pretectal nucleus, nucleus of the posterior commissure, superior colliculus, inferior colliculus, periaqueductal grey, substantia nigra and raphe nuclei. Few notable changes in the profile of GATA3 expression occurred over this time period. As postnatal development progressed, however, GATA3 expression weakened, and was maintained in only a few regions of the adult CNS. Throughout the brain, we found that GATA3-expressing cells were NeuN-positive, and no colocalization with glial fibrillary acidic protein (GFAP) was observed. In the substantia nigra, GATA3 was exclusively expressed in cells of the reticulate part and some of which were found to be GABAergic. This study presents a comprehensive overview of GATA3 expression in the CNS throughout postnatal life, and the dynamics that we observed provide insights for further investigations of the roles of GATA3 in postnatal development and the maintenance of the mature CNS.


Subject(s)
Central Nervous System/growth & development , Central Nervous System/metabolism , GATA3 Transcription Factor/metabolism , Gene Expression Regulation, Developmental/physiology , Age Factors , Animals , Animals, Newborn , Central Nervous System/cytology , GATA3 Transcription Factor/genetics , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Mice , Mice, Inbred C57BL , Neurons/metabolism , RNA, Messenger/metabolism , Tyrosine 3-Monooxygenase/metabolism
14.
J Comp Neurol ; 509(4): 341-55, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18512225

ABSTRACT

The expression profile of Lim homeodomain transcription factor Lmx1b in the mouse brain was investigated at different postnatal stages by immunohistochemistry and in situ hybridization. At postnatal day (P) 7, many Lmx1b-expressing neurons were found in the posterior hypothalamic area, supramammillary nucleus, ventral premammillary nucleus, and subthalamic nucleus. In the midbrain, numerous Lmx1b-expressing neurons were present in the substantia nigra pars compacta and ventral tegmental area. In the hindbrain, Lmx1b-expressing neurons were primarily observed in the raphe nuclei, parabrachial nuclei, principal sensory trigeminal nucleus, nucleus of the solitary tract, and laminae I-II of the medullary dorsal horn as well as spinal dorsal horn. Although expression levels diminished as postnatal life progressed, persistent expression throughout the first year of life was observed in many of these regions. In contrast, Lmx1b was present in a few brain regions (e.g., principal sensory trigeminal nucleus) only in early life with expression expiring by P60. Lmx1b was observed in dopaminergic neurons in the midbrain and serotonergic neurons in the hindbrain, as determined by double labeling with specific markers. In addition, we found that Lmx1b-expressing neurons are not GABAergic, and Lmx1b was colocalized with Tlx3 in the parabrachial nuclei, principal sensory trigeminal nucleus, nucleus of the solitary tract. as well as the medullary and spinal dorsal horns, suggesting that Lmx1b-expressing cells in these areas are excitatory neurons. Our data suggest that Lmx1b is involved in the postnatal maturation of certain types of neurons and maintenance of their normal functions in the adult brain.


Subject(s)
Brain/physiology , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Neurons/physiology , Spinal Cord/physiology , Transcription Factors/genetics , Animals , Animals, Newborn , Brain/growth & development , Hypothalamus/growth & development , Hypothalamus/physiology , In Situ Hybridization , LIM-Homeodomain Proteins , Medulla Oblongata/growth & development , Medulla Oblongata/physiology , Mice , Mice, Inbred C57BL , Pons/growth & development , Pons/physiology , Spinal Cord/growth & development , Thalamus/growth & development , Thalamus/physiology
15.
Ann Clin Lab Sci ; 37(4): 356-61, 2007.
Article in English | MEDLINE | ID: mdl-18000293

ABSTRACT

Previous studies have shown that labile zinc and inflammatory mediators participate in many pathophysiological processes. The present study investigated the effects of traumatic brain injury (TBI) on the levels of labile zinc and certain proinflammatory factors in rat lung. Male Wistar rats were randomly assigned to 7 groups as follows: normal group, group with sham operation, and TBI groups that were sacrificed respectively at 1, 6, 24, and 72 hr, and on day 7 post-injury. Pulmonary labile zinc, tumor necrosis factor alpha (TNF-alpha), interleukin (IL)-8, and wet/dry weight ratio were measured at the specified time intervals. TBI caused a gradual increase of pulmonary labile zinc as demonstrated by fluorescence staining with Zinpyr-4 (ZP4). The levels of TNF-alpha and IL-8 and the lung wet/dry weight ratios were higher in the TBI groups compared to the normal and sham-operated groups (p <0.05). There were highly positive correlations between the intensity of ZP4 fluorescence and the pulmonary levels of TNF-alpha and IL-8. The results suggest that TBI induces rapid increases of labile zinc and inflammatory mediators in lung, which may participate in the pathogenesis of acute lung injury.


Subject(s)
Brain Injuries/metabolism , Cytokines/metabolism , Lung/metabolism , Zinc/metabolism , Animals , Disease Progression , Fluorescent Dyes/metabolism , Homeostasis , Interleukin-8/metabolism , Lung/pathology , Male , Organ Size , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...