Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Neuroscience ; 252: 262-76, 2013 Nov 12.
Article in English | MEDLINE | ID: mdl-23892008

ABSTRACT

Focal malformations of cortical development (FMCD) are highly associated with several neurological disorders including intractable epilepsy and neurocognitive disabilities. Over the past decade, several FMCD subtypes have been linked to hyperactivation of the mammalian target of rapamycin (mTOR) signaling cascade. In view of the roles that mTOR plays in cell proliferation, size, motility, and stem cell phenotype, many of the features of FMCD such as cytomegaly, disorganized lamination, and expression of stem cell markers can be explained by enhanced mTOR signaling. FMCD result from several distinct and fascinating molecular mechanisms including biallelic gene inactivation, somatic mutation, and potentially, viral infection. These mechanisms have been directly linked to mTOR activation. Perhaps most compelling, pharmacological inhibition of mTOR has been implemented successfully in clinical trials for select FMCD and provides a new vista for treatment.


Subject(s)
Malformations of Cortical Development/genetics , Malformations of Cortical Development/metabolism , Signal Transduction/physiology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Animals , Humans
3.
Epilepsy Res ; 78(1): 7-21, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18023148

ABSTRACT

Cortical tubers and subependymal giant cell tumors (SGCT) are two major cerebral lesions associated with tuberous sclerosis complex (TSC). In the present study, we investigated immunocytochemically the inflammatory cell components and the induction of two major pro-inflammatory pathways (the interleukin (IL)-1beta and complement pathways) in tubers and SGCT resected from TSC patients. All lesions were characterized by the prominent presence of microglial cells expressing class II-antigens (HLA-DR) and, to a lesser extent, the presence of CD68-positive macrophages. We also observed perivascular and parenchymal T lymphocytes (CD3(+)) with a predominance of CD8(+) T-cytotoxic/suppressor lymphoid cells. Activated microglia and reactive astrocytes expressed IL-1beta and its signaling receptor IL-1RI, as well as components of the complement cascade, such as C1q, C3c and C3d. Albumin extravasation, with uptake in astrocytes, was observed in both tubers and SGCT, suggesting that alterations in blood brain barrier permeability are associated with inflammation in TSC-associated lesions. Our findings demonstrate a persistent and complex activation of inflammatory pathways in cortical tubers and SGCT.


Subject(s)
Brain Neoplasms/complications , Cerebral Cortex/pathology , Giant Cell Tumors/complications , Inflammation/etiology , Tuberous Sclerosis/complications , Adolescent , Adult , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , CD3 Complex/metabolism , Child , Child, Preschool , Female , Glial Fibrillary Acidic Protein/metabolism , HLA-DR Antigens/metabolism , Humans , Infant , Interleukin-1beta/metabolism , Male , Neuroglia/metabolism , Neurons/metabolism , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Proteins
4.
Neuropathol Appl Neurobiol ; 33(4): 455-70, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17617874

ABSTRACT

Hemimegalencephaly (HMEG) is a malformation of cortical development characterized by unilateral enlargement of the cerebral hemisphere, severe architectural and cellular abnormalities and association with intractable epilepsy. HMEG may represent an isolated lesion of the central nervous system, but may also be associated with several neurocutaneous syndromes. In the present study we discuss the neuropathological findings of two autopsy cases of HMEG associated with linear naevus sebaceous syndrome. Both cases showed the presence of linear naevus sebaceous on extensive areas of the face. The neurochemical profile of the glial and neuronal components in the affected hemisphere was determined using immunocytochemical markers and was compared with the unaffected contralateral hemisphere and normal control tissue. The observed cytomegalic neurones expressed receptors for distinct neurotransmitters, neuropeptides and growth factors. Analysis of components of the phosphoinositide 3-kinase pathway revealed expression of phospho-S6 ribosomal protein in cytomegalic neurones. Autopsy findings confirm the complexity of the histologic phenotypic manifestations in HMEG and proved useful in determining the spectrum of cytoarchitectural and neurochemical abnormalities, underlying the molecular pathogenesis and epileptogenesis of this brain malformation.


Subject(s)
Cerebral Cortex/abnormalities , Cerebral Cortex/pathology , Adolescent , Autopsy , Brain Chemistry/physiology , Cell Count , Functional Laterality/physiology , Humans , Immunohistochemistry , Infant, Newborn , Male , Nerve Tissue Proteins/metabolism , Neurons/pathology , Neurons/ultrastructure , Neuropeptide Y/metabolism , Nevus/pathology , Receptors, Glutamate/metabolism , Seizures/etiology , Syndrome , Tissue Fixation , Vascular Endothelial Growth Factor A/metabolism
5.
Neurology ; 62(6 Suppl 3): S9-13, 2004 Mar 23.
Article in English | MEDLINE | ID: mdl-15037672

ABSTRACT

Cortical dysplasias (CDs) increasingly are recognized as pathologic substrates in patients with medically intractable epilepsy. Several studies have demonstrated the intrinsic epileptogenicity of these lesions, but the cellular and molecular mechanisms responsible for seizure initiation remain unknown. The increased availability of surgically resected neocortical tissue has provided the opportunity for direct histopathologic and electrocorticographic correlations. Moreover, the description of various animal models of CDs allowed the testing of various mechanistic hypotheses. It is likely that the mechanisms of epileptogenicity in CDs are multifactorial. In this article, the authors summarize current knowledge of the molecular and cellular mechanisms of epileptogenicity in focal CDs based on human and animal data. In particular, they focus on the roles of glutamate (NMDA and alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid) and gamma-aminobutyric acid receptors identified in animal models and resected human neocortex.


Subject(s)
Brain Diseases/complications , Epilepsy/etiology , Nervous System Malformations/complications , Animals , Brain Diseases/physiopathology , Electrophysiology , Epilepsy/physiopathology , Humans , Nervous System Malformations/physiopathology , Neurotransmitter Agents/physiology , Receptors, Glutamate/metabolism
6.
Am J Pathol ; 159(4): 1541-54, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11583980

ABSTRACT

Neurotrophins and their receptors modulate cerebral cortical development. Tubers in the tuberous sclerosis complex (TSC) are characterized histologically by disorganized cortical cytoarchitecture and thus, we hypothesized that expression of neurotrophin mRNAs and proteins might be altered in tubers. Using in situ transcription and mRNA amplification to probe cDNA arrays, we found that neurotrophin-3 (NT3) and trkB mRNA expression were reduced whereas neurotrophin-4 (NT4) and trkC mRNA expression were increased in whole tuber sections. Alterations in mRNA abundance were defined in single microdissected dysplastic neurons (DNs) and giant cells (GCs). NT3 mRNA expression was reduced in GCs and trkB mRNA expression was reduced in DNs. NT4 mRNA expression was increased in DNs and trkC mRNA expression was increased in both DNs and GCs. In three patients, TSC2 locus mutations were confirmed and the mean tuberin mRNA expression levels was reduced across all nine cases. Consistent with these observations, NT3 mRNA expression was reduced but trkC mRNA expression was increased in vitro in human NTera2 neurons (NT2N) transfected with a tuberin antisense construct that reduced tuberin expression. Western analysis of tuber homogenates and computer-assisted densitometry of immunolabeled sections confirmed the neurotrophin mRNA expression data in whole sections and single neurotrophin immunoreactive cells. We conclude that alterations in NT4/trkB and NT3/trkC expression may contribute to tuber formation during brain development as downstream effects of the hamartin and tuberin pathway in TSC.


Subject(s)
Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Nerve Growth Factors/metabolism , Tuberous Sclerosis/metabolism , Tuberous Sclerosis/pathology , Adolescent , Adult , Antisense Elements (Genetics)/genetics , Blotting, Western , Cell Line , Child , Child, Preschool , Female , Gene Amplification , Gene Expression , Genotype , Giant Cells/physiology , Humans , Immunohistochemistry , Infant , Male , Middle Aged , Nerve Growth Factors/genetics , Neurons/pathology , Neurons/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transfection , Tuberous Sclerosis/genetics , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins
7.
Hum Mol Genet ; 10(17): 1775-83, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11532987

ABSTRACT

Periventricular heterotopia (PH) is a human neuronal migration disorder in which many neurons destined for the cerebral cortex fail to migrate. Previous analysis showed heterozygous mutations in the X-linked gene filamin 1 (FLN1), but examined only the first six (of 48) coding exons of the gene and hence did not assess the incidence and functional consequences of FLN1 mutations. Here we perform single-strand conformation polymorphism (SSCP) analysis of FLN1 throughout its entire coding region in six PH pedigrees, 31 sporadic female PH patients and 24 sporadic male PH patients. We detected FLN1 mutations by SSCP in 83% of PH pedigrees and 19% of sporadic females with PH. Moreover, no PH females (0/7 tested) with atypical radiographic features showed FLN1 mutations, suggesting that other genes may cause atypical PH. Surprisingly, 2/24 males analyzed with PH (9%) also carried FLN1 mutations. Whereas FLN1 mutations in PH pedigrees caused severe predicted loss of FLN1 protein function, both male FLN1 mutations were consistent with partial loss of function of the protein. Moreover, sporadic female FLN1 mutations associated with PH appear to cause either severe or partial loss of function. Neither male could be shown to be mosaic for the FLN1 mutation in peripheral blood lymphocytes, suggesting that some neurons in the intact cortex of PH males may be mutant for FLN1 but migrate adequately. These results demonstrate the sensitivity and specificity of DNA testing for FLN1 mutations and have important functional implications for models of FLN1 protein function in neuronal migration.


Subject(s)
Abnormalities, Multiple/genetics , Cerebral Cortex/abnormalities , Cerebral Ventricles/abnormalities , Contractile Proteins/genetics , Microfilament Proteins/genetics , Sex Chromosome Aberrations , X Chromosome , Aging , Cerebral Cortex/pathology , Cerebral Ventricles/pathology , DNA Mutational Analysis , DNA Primers , Female , Filamins , Humans , Magnetic Resonance Imaging , Male , Neurons/pathology , Phenotype , Polymorphism, Single-Stranded Conformational , Sex Characteristics
8.
Acta Neuropathol ; 102(2): 141-8, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11563628

ABSTRACT

Focal cortical dysplasia (FCD) is characterized by disorganized cerebral cortical cytoarchitecture. Increased expression of several intermediate filament (IF) proteins such as neurofilament, vimentin, alpha-internexin, and nestin observed in dysplastic "balloon" neurons (DN) may contribute to disrupted cortical lamination. We hypothesized that increased IF protein expression results from enhanced IF gene transcription within dysplastic neurons. We used a novel strategy to evaluate IF mRNA expression in three FCD specimens from medically intractable epilepsy patients. Poly(A) mRNA was amplified (aRNA) from single microdissected DN, morphologically normal neurons at the margin of the FCD resection, morphologically normal neurons in non-FCD cortex from epilepsy patients, and normal control neurons. Radiolabeled aRNA from single neurons was used to probe cDNA arrays containing the low (NFL), medium (NFM) and high (NFH) molecular weight neurofilament isoform, alpha-internexin, desmin, vimentin, peripherin (PRPH), nestin, and glial fibrillary acidic protein (GFAP) cDNAs. Hybridization intensity of aRNA-cDNA hybrids was used to quantify relative IF abundance. Increased expression of nestin, alpha-internexin, PRPH, vimentin, NFL, NFM, and NFH mRNAs was found in DN when compared with the three control neuronal subtypes. Desmin and GFAP mRNAs were not detected in any cell types. Expression of PRPH mRNA and protein in select DN was confirmed by reverse transcription-polymerase chain reaction and immunohistochemistry. We conclude that aberrant expression of IF proteins in FCD likely results from enhanced transcription of IF genes in dysplastic neurons and propose that future analysis of transcriptional elements that regulate IF expression be evaluated in FCD.


Subject(s)
Cell Movement/genetics , Cerebral Cortex/abnormalities , Cerebral Cortex/metabolism , Epilepsy/genetics , Intermediate Filament Proteins/genetics , Membrane Glycoproteins , Nervous System Malformations/genetics , Neurons/metabolism , Transcription, Genetic/genetics , Adolescent , Adult , Carrier Proteins/genetics , Cerebral Cortex/pathology , Desmin/genetics , Dissection/methods , Epilepsy/metabolism , Epilepsy/pathology , Female , Gene Expression Regulation, Developmental/physiology , Glial Fibrillary Acidic Protein/genetics , Humans , Intermediate Filament Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nervous System Malformations/metabolism , Nervous System Malformations/pathology , Nestin , Neurofilament Proteins/genetics , Neurofilament Proteins/metabolism , Neurons/pathology , Oligonucleotide Array Sequence Analysis , Pathology/methods , Peripherins , RNA, Messenger/metabolism , Vimentin/genetics
9.
J Neurosci Res ; 65(2): 150-64, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11438984

ABSTRACT

Ezrin, radixin, and moesin (ERM proteins), as well as the neurofibromatosis 2 (NF2) tumor suppressor merlin/schwannomin, all belong to the protein 4.1 family, yet only merlin is a tumor suppressor in Schwann cells. To gain insight into the possible functions of ERM proteins in Schwann cells, we examined their localization in peripheral nerve, because we have previously shown that merlin is found in paranodes and in Schmidt-Lanterman incisures. All three ERM proteins were highly expressed in the microvilli of myelinating Schwann cells that surround the nodal axolemma as well as in incisures and cytoplasmic puncta in the vicinity of the node. In all of these locations, ERM proteins were colocalized with actin filaments. In contrast, ERM proteins did not surround nodes in the CNS. The colocalization of ERM proteins with actin indicates that they have functions different from those of merlin in myelinating Schwann cells.


Subject(s)
Blood Proteins/metabolism , Cytoskeletal Proteins/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Microvilli/metabolism , Myelin Sheath/metabolism , Phosphoproteins/metabolism , Potassium Channels, Voltage-Gated , Schwann Cells/metabolism , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Animals , Ankyrins/metabolism , Antibody Specificity/physiology , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Central Nervous System/metabolism , Central Nervous System/ultrastructure , Immunohistochemistry , Kv1.1 Potassium Channel , Kv1.2 Potassium Channel , Microscopy, Electron , Microvilli/ultrastructure , Myelin-Associated Glycoprotein/metabolism , Neurofilament Proteins/metabolism , Peripheral Nerves/metabolism , Peripheral Nerves/ultrastructure , Potassium Channels/metabolism , RNA, Messenger/metabolism , Ranvier's Nodes/metabolism , Ranvier's Nodes/ultrastructure , Rats , Schwann Cells/ultrastructure , Sciatic Nerve/metabolism , Sciatic Nerve/ultrastructure , Sodium Channels/metabolism
10.
J Exp Clin Cancer Res ; 20(1): 5-10, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11370829

ABSTRACT

Glioblastoma multiforme (GBM) has associated with it one of the poorest prognoses among brain tumors. Postoperative seizures and the side effects of anticonvulsants, routinely given for prophylactic purposes, add to patient morbidity. The primary goal of this study was to determine who, of those undergoing craniotomy for GBM resection, is at risk for epilepsy. We studied 72 consecutive patients who underwent craniotomy and palliative resection for GBM. Twenty-nine presented with seizures and 17 had postoperative seizures. All patients were treated with a postoperative anticonvulsant for at least six months; anticonvulsants were continued longer if there was a postoperative seizure. Patient factors examined for an association with risk for postoperative seizure included age, sex, tumor size, tumor location, adjuvant therapy, postoperative complications and history of preoperative seizures. The majority of patients with no prior seizure history and who seized postoperatively had their first seizure after withdrawal from their anticonvulsant medication. All, but one, of the patients with both pre- and postoperative seizures had their first postoperative seizure while still on anticonvulsants. Smaller tumor size and frontal resection were associated with an increased risk of postoperative seizures. Our data suggests that those who do not present with seizures and undergo GBM resection may still be prone to seize but more easily protected from postoperative seizures with anticonvulsant therapy than patients who present with seizures; resection of frontal tumors and smaller tumors seemed to indicate an increased risk for postoperative seizures.


Subject(s)
Brain Neoplasms/surgery , Craniotomy/adverse effects , Epilepsy/epidemiology , Glioblastoma/surgery , Postoperative Complications/epidemiology , Anticonvulsants/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Chemotherapy, Adjuvant , Combined Modality Therapy , Epilepsy/drug therapy , Epilepsy/etiology , Female , Genetic Therapy , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Palliative Care , Postoperative Complications/physiopathology , Prognosis , Retrospective Studies , Seizures/drug therapy , Seizures/epidemiology , Seizures/etiology , Time Factors
11.
Neurology ; 56(7): 906-13, 2001 Apr 10.
Article in English | MEDLINE | ID: mdl-11294928

ABSTRACT

OBJECTIVE: Focal cortical dysplasia is characterized by disorganized cortical lamination, dysplastic and heterotopic neurons, and an association with epilepsy. The contribution that dysplastic and heterotopic neurons make to epileptogenesis in focal cortical dysplasia is unknown and the phenotype of these cells may be distinct. The authors hypothesized that the expression of genes encoding glutamatergic (glutamate [GluR] and N-methyl-D-aspartate NMDA receptors [NR]) and gamma-aminobutyric acid A receptor (GABA(A)R) subunits is distinct in dysplastic and heterotopic neurons and that changes in receptor gene expression could be defined in a cell-specific pattern. METHODS: Single immunohistochemically labeled dysplastic and heterotopic neurons were microdissected from human focal cortical dysplasia specimens obtained during epilepsy surgery. Pyramidal neurons were microdissected from postmortem control cortex and from temporal cortex without dysplasia resected during temporal lobectomy. Poly (A) messenger RNA (mRNA) from single neurons was amplified, radiolabeled, and used to probe complementary DNA (cDNA) arrays containing GluR(1-6), NR(1A,1B), NR(2A-D), and GABA(A)Ralpha(1-6), and -Rbeta(1-3) subunit cDNAS: The relative hybridization intensities of each mRNA-cDNA hybrid were quantified by phosphorimaging. RESULTS: GluR, NR, and GABA(A)R subunit mRNA expression did not differ between control neurons and nondysplastic epilepsy specimens. Expression of GluR(4), NR(2B), and NR(2C) subunit mRNA was increased, and NR(2A) and GABA(A)Rbeta(1) subunit mRNA was decreased in dysplastic compared with pyramidal and heterotopic neurons. In contrast, GABA(A)Ralpha(1), -Ralpha(2), and -Rbeta(2) as well as GluR(1) mRNA levels were reduced in both dysplastic and heterotopic neurons. CONCLUSIONS: Differential expression of GluR, NR, and GABA(A)R mRNA in dysplastic and heterotopic neurons demonstrates cell specific gene transcription changes in focal cortical dysplasia. These results suggest that dysplastic and heterotopic neurons may be pharmacologically distinct and make differential contributions epileptogenesis in focal cortical dysplasia.


Subject(s)
Brain Diseases/pathology , Neurons/pathology , RNA, Messenger/analysis , Receptors, GABA-A/genetics , Receptors, Glutamate/genetics , Adolescent , Adult , Brain Diseases/genetics , Dissection/methods , Female , Humans , Immunohistochemistry , Male , Middle Aged
12.
Ann Neurol ; 49(1): 67-78, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11198298

ABSTRACT

The molecular pharmacologic basis of epileptogenesis in cortical tubers in the tuberous sclerosis complex is unknown. Altered transcription of genes encoding glutamatergic and gamma-aminobutyric acid (GABA)-ergic receptors and uptake sites may contribute to seizure initiation and may occur selectively in dysplastic neurons and giant cells. Arrays containing GABA A (GABAAR), GluR, NMDA receptor (NR) subunits, GAD65, the vesicular GABA transporter (VGAT), and the neuronal glutamate transporter (EAAC1) cDNAs were probed with amplified poly (A) mRNA from tubers or normal neocortex to identify changes in gene expression. Increased levels of EAAC1, and NR2B and 2D subunit mRNAs and diminished levels of GAD65, VGAT, GluR1, and GABAAR alpha1 and alpha2 were observed in tubers. Ligand-binding experiments in frozen tuber homogenates demonstrated an increase in functional NR2B-containing receptors. Arrays were then probed with poly (A) mRNA from single, microdissected dysplastic neurons, giant cells, or normal neurons (n = 30 each). Enhanced expression of GluR 3, 4, and 6 and NR2B and 2C subunit mRNAs was noted in the dysplastic neurons, whereas only the NR2D mRNA was upregulated in giant cells. GABAAR alpha1 and alpha2 mRNA levels were reduced in both dysplastic neurons and giant cells compared to control neurons. Differential expression of GluR, NR, and GABAAR mRNAs in tubers reflects cell-specific changes in gene transcription that argue for a distinct molecular phenotype of dysplastic neurons and giant cells and suggests that dysplastic neurons and giant cells make differential contributions to epileptogenesis in the tuberous sclerosis complex.


Subject(s)
Brain/pathology , Giant Cells/pathology , Glutamic Acid/analysis , Neurons/pathology , Receptors, GABA/analysis , Tuberous Sclerosis/pathology , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Immunohistochemistry , Male , Middle Aged
13.
Curr Protoc Neurosci ; Chapter 5: Unit 5.3, 2001 May.
Article in English | MEDLINE | ID: mdl-18428502

ABSTRACT

This unit presents a method for the amplification of poly(A)(+) mRNA extracted from the cytoplasm of a single cell. After cDNA is synthesized from the mRNA, it is made double stranded, denatured, and reverse transcribed to yield antisense RNA (aRNA). Another round of amplification results in a relatively large amount of aRNAs in essentially the same proportion as in the starting mRNA population. RNA amplification protocols can be used for many purposes, including generation of disease expression profiles, making of cDNA libraries, and generation of diagnostics and therapeutics for disease. An alternate protocol is used to amplify RNAs from single neurons in fixed tissue specimens. Support protocols gives instructions for reverse northern analysis, which allows analysis of the presence or absence and relative levels of mRNA expression in selected cells, and a convenient method to assess the RNA content in fixed tissue sections using the fluorescent dye acridine orange (which binds single-stranded nucleic acids).


Subject(s)
Central Nervous System/cytology , Central Nervous System/physiology , Cytoplasm/genetics , RNA, Messenger/genetics , Animals , Cells, Cultured , Central Nervous System/chemistry , Cytoplasm/chemistry , Humans , Organ Culture Techniques , RNA, Messenger/analysis
15.
Proc Natl Acad Sci U S A ; 97(21): 11545-50, 2000 Oct 10.
Article in English | MEDLINE | ID: mdl-11027353

ABSTRACT

The selective subcellular localization of mRNAs to dendrites and the recent demonstration of local protein synthesis have highlighted the potential role of postsynaptic sites in modulation of cell-cell communication. We show that epitope-tagged subunit 2 of the ionotopic glutamate receptor, GluR2, mRNA transfected into isolated hippocampal neuronal dendrites is translated in response to pharmacologic stimulation. Further, confocal imaging of N-terminally labeled GluR2 reveals that the newly synthesized GluR2 protein can integrate into the dendritic membrane with the N terminus externally localized. These data demonstrate that integral membrane proteins can be synthesized in dendrites and can locally integrate into the cell membrane.


Subject(s)
Dendrites/metabolism , Membrane Proteins/biosynthesis , Neurons/metabolism , Receptors, Glutamate/biosynthesis , Amino Acid Sequence , Animals , Base Sequence , DNA Primers , Endoplasmic Reticulum, Rough/metabolism , Genes, myc , Golgi Apparatus/metabolism , Immunohistochemistry , Membrane Proteins/genetics , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, Glutamate/genetics , Transfection
16.
J Neurosci ; 20(13): 4821-8, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10864939

ABSTRACT

The cerebral cortex is selectively vulnerable to cell death after traumatic brain injury (TBI). We hypothesized that the ratio of mRNAs encoding proteins important for cell survival and/or cell death is altered in individual damaged neurons after injury that may contribute to the cell's fate. To investigate this possibility, we used amplified antisense mRNA (aRNA) amplification to examine the relative abundance of 31 selected candidate mRNAs in individual cortical neurons with fragmented DNA at 12 or 24 hr after lateral fluid percussion brain injury in anesthetized rats. Only pyramidal neurons characterized by nuclear terminal deoxynucleotidyl transferase-mediated biotinylated dUTP nick end labeling (TUNEL) reactivity with little cytoplasmic staining were analyzed. For controls, non-TUNEL-positive neurons from the cortex of sham-injured animals were obtained and subjected to aRNA amplification. At 12 hr after injury, injured neurons exhibited a decrease in the relative abundance of specific mRNAs including those encoding for endogenous neuroprotective proteins. By 24 hr after injury, many of the mRNAs altered at 12 hr after injury had returned to baseline (sham-injured) levels except for increases in caspase-2 and bax mRNAs. These data suggest that TBI induces a temporal and selective alteration in the gene expression profiles or "molecular fingerprints" of TUNEL-positive neurons in the cerebral cortex. These patterns of gene expression may provide information about the molecular basis of cell death in this region after TBI and may suggest multiple avenues for therapeutic intervention.


Subject(s)
Apoptosis , Brain Injuries/physiopathology , Cerebral Cortex/physiopathology , Nerve Tissue Proteins/genetics , Neurons/pathology , Neurons/physiology , Animals , Brain Injuries/genetics , Brain Injuries/pathology , Caspase 2 , Caspases/genetics , Cell Death , Cerebral Cortex/pathology , Cyclic AMP Response Element-Binding Protein/genetics , Gene Expression Regulation , Genes, fos , In Situ Nick-End Labeling , Inflammation , Male , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Antisense , Rats , Rats, Sprague-Dawley , Receptors, AMPA/genetics , Receptors, N-Methyl-D-Aspartate/genetics , gamma-Aminobutyric Acid/metabolism
17.
J Neurovirol ; 6 Suppl 1: S61-9, 2000 May.
Article in English | MEDLINE | ID: mdl-10871767

ABSTRACT

APJ is a recently described seven-transmembrane (7TM) receptor that is abundantly expressed in the central nervous system (CNS). This suggests an important role for APJ in neural development and/or function, but neither its cellular distribution nor its function have been defined. APJ can also serve as a co-receptor with CD4 for fusion and infection by some strains of human immunodeficiency virus (HIV-1) in vitro, suggesting a role in HIV neuropathogenesis if it were expressed on CD4-positive CNS cells. To address this, we examined APJ expression in cultured neurons, astrocytes, oligodendrocytes, microglia and monocyte-derived macrophages utilizing both immunocytochemical staining with a polyclonal anti-APJ antibody and RT - PCR. We also analyzed the ability of a recently identified APJ peptide ligand, apelin, to induce calcium elevations in cultured neural cells. APJ was expressed at a high level in neurons and oligodendrocytes, and at lower levels in astrocytes. In contrast, APJ was not expressed in either primary microglia or monocyte-derived macrophages. Several forms of the APJ peptide ligand induced calcium elevations in neurons. Thus, APJ is selectively expressed in certain CNS cell types and mediates intracellular signals in neurons, suggesting that APJ may normally play a role in signaling in the CNS. However, the absence of APJ expression in microglia and macrophages, the prinicpal CD4-positive cell types in the brain, indicates that APJ is unlikely to mediate HIV-1 infection in the CNS.


Subject(s)
Astrocytes/metabolism , Macrophages/metabolism , Microglia/metabolism , Neurons/metabolism , Oligodendroglia/metabolism , Receptors, Dopamine D2/metabolism , Receptors, G-Protein-Coupled , Adult , Amino Acid Sequence , Antibodies , Apelin Receptors , Blotting, Southern , Brain/cytology , Brain/metabolism , Calcium/metabolism , Fetus , Fluorescent Antibody Technique, Indirect , Humans , Molecular Sequence Data , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
18.
Neurology ; 53(7): 1384-90, 1999 Oct 22.
Article in English | MEDLINE | ID: mdl-10534239

ABSTRACT

OBJECTIVE: To outline recent developments in the neurobiology of the tuberous sclerosis complex (TSC). BACKGROUND: TSC may be associated with neuropsychiatric disorders including epilepsy, mental retardation, and autism. The uncontrolled growth of subependymal giant cell astrocytomas may lead to hydrocephalus and death. The recent identification of mutations in two genes (TSC1 and TSC2) that cause TSC has led to rapid progress in understanding the molecular and cellular pathogenesis of this disorder. How distinct mutations lead to the varied clinical phenotype of TSC is under intense investigation. RESULTS: We report the recent diagnostic criteria for TSC and provide an overview of the molecular genetics, molecular pathophysiology, and neuropathology of TSC. Important diagnostic criteria for TSC include facial angiofibromas, ungual fibromas, retinal hamartomas, and cortical tubers. Both familial and sporadic TSC cases occur. Approximately 50% of TSC families show genetic linkage to TSC1 and 50% to TSC2. Among sporadic TSC cases, mutations in TSC2 are more frequent and often accompanied by more severe neurologic deficits. Multiple mutational subtypes have been identified in the TSC1 and TSC2 genes. The TSC1 (chromosome 9) and TSC2 (chromosome 16) genes encode distinct proteins, hamartin and tuberin, respectively, which are widely expressed in the brain and may interact as part of a cascade pathway that modulates cellular differentiation, tumor suppression, and intracellular signaling. Tuberin has a GTPase activating protein-related domain that may contribute to a role in cell cycle passage and intracellular vesicular trafficking. CONCLUSION: Identification of tuberous sclerosis complex (TSC) gene mutations has fostered understanding of how brain lesions in TSC are formed. Further characterization of the roles of hamartin and tuberin will provide potential therapeutic avenues to treat seizures, mental retardation, and tumor growth in TSC.


Subject(s)
Neurobiology/trends , Tuberous Sclerosis/diagnosis , Tuberous Sclerosis/genetics , Brain/pathology , Humans , Magnetic Resonance Imaging , Retina/pathology , Tuberous Sclerosis/pathology
19.
Methods Enzymol ; 303: 3-18, 1999.
Article in English | MEDLINE | ID: mdl-10349635

ABSTRACT

Phenotypic characterization of cells in conjunction with single-cell mRNA analysis, which yields information regarding expression of multiple genes in individual neurons, facilitates a detailed and comprehensive view of neuronal cell biology. More specifically, the aRNA amplification method has provided an approach to analyze mRNA levels in single cells that have been phenotypically characterized on the basis of electrophysiology, morphology, and/or protein expression. In this way, relative mRNA abundances can be directly assayed from a well-defined population of neurons. The concept of expression profiling led to the development of robotics methods for arraying thousands of cDNAs on microarrays. These cDNA arrays can be screened with labeled aRNA or cDNA to generate a molecular fingerprint of a specific cell type, disease state, or therapeutic efficacy. A broad view of how gene expression is altered in single neurons affected by a particular disease process may provide clues to pathogenetic disease mechanisms or avenues for therapeutic interventions. The use of mRNA profiles to produce diagnostics and therapeutics is called transcript-aided drug design (TADD). When coupled with single-cell resolution, TADD promises to be an important tool in diagnosis of disease states, as well as provide a blueprint on which to develop therapeutic strategies. For example, mRNA abundances in an individual diseased cell may increase, decrease, or remain constant, and thus it is possible that a pharmaceutical alone or in combination with other drugs may be specifically designed to restore mRNA abundances to a normal state. Alternatively, if functional protein levels parallel the mRNA level changes, then drugs targeting the function of the proteins translated from these altered mRNAs may prove to be therapeutic. One promise of such an approach is that information about mRNA abundances that are altered in a diseased cell may provide new therapeutic indications for existing drugs. For example, if the abundance of mRNA for the beta-adrenergic receptor is altered as shown by the microarrays for a particular disease, already available adrenergic receptor agonists or antagonists that had not previously been used in this particular disease paradigm may prove to be therapeutically efficacious. The expression profile of a given cell is a measure of the potential for protein expression. Proteins are generally the functional entities within cells and differences in protein function often result in disease. The ability to monitor the coordinate changes in gene expression, in single phenotypically identified cells, that correlate with disease will provide unique insight into the expressed genetic variability of cells and will likely furnish unforeseen insight into the underlying cellular mechanisms that produce disease etiology.


Subject(s)
DNA, Complementary/isolation & purification , Hippocampus/metabolism , Neurons/metabolism , RNA, Messenger/genetics , Animals , Base Sequence , Cells, Cultured , DNA, Complementary/biosynthesis , Dendrites/metabolism , Drug Design , Humans , Nucleic Acid Amplification Techniques , Promoter Regions, Genetic , RNA, Antisense/genetics , Subcellular Fractions/chemistry , Subcellular Fractions/metabolism , Transcription, Genetic
20.
Ann Neurol ; 45(2): 174-81, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9989619

ABSTRACT

The sequestration of RNA in Alzheimer's disease (AD) senile plaques (SPs) and the production of intraneuronal amyloid-beta peptides (Abeta) prompted analysis of the mRNA profile in single immunocytochemically identified SPs in sections of AD hippocampus. By using amplified RNA expression profiling, polymerase chain reaction, and in situ hybridization, we assessed the presence and abundance of 51 mRNAs that encode proteins implicated in the pathogenesis of AD. The mRNAs in SPs were compared with those in individual CA1 neurons and the surrounding neuropil of control subjects. The remarkable demonstration here, that neuronal mRNAs predominate in SPs, implies that these mRNAs are nonproteinaceous components of SPs, and, moreover, that mRNAs may interact with Abeta protein and that SPs form at sites where neurons degenerate in the AD brain.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Plaque, Amyloid/metabolism , RNA, Messenger/analysis , Aged , Blotting, Northern , Female , Humans , Immunohistochemistry , In Situ Hybridization , Male , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...