Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Cell Biol ; 221(7)2022 07 04.
Article in English | MEDLINE | ID: mdl-35579602

ABSTRACT

Neuregulins (NRGs) are EGF-like ligands associated with cognitive disorders. Unprocessed proNRG3 is cleaved by BACE1 to generate the mature membrane-bound NRG3 ligand, but the subcellular site of proNRG3 cleavage, mechanisms underlying its transport into axons, and presynaptic accumulation remain unknown. Using an optogenetic proNRG3 cleavage reporter (LA143-NRG3), we investigate the spatial-temporal dynamics of NRG3 processing and sorting in neurons. In dark conditions, unprocessed LA143-NRG3 is retained in the trans-Golgi network but, upon photoactivation, is cleaved by BACE1 and released from the TGN. Mature NRG3 then emerges on the somatodendritic plasma membrane from where it is re-endocytosed and anterogradely transported on Rab4+ vesicles into axons via transcytosis. By contrast, the BACE1 substrate APP is sorted into axons on Rab11+ vesicles. Lastly, by a mechanism we denote "trans-synaptic retention," NRG3 accumulates at presynaptic terminals by stable interaction with its receptor ErbB4 on postsynaptic GABAergic interneurons. We propose that trans-synaptic retention may account for polarized expression of other neuronal transmembrane ligands and receptors.


Subject(s)
Axons , Neuregulins , Receptor, ErbB-4 , Transcytosis , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Axons/metabolism , HEK293 Cells , Humans , Ligands , Mice , Neuregulins/genetics , Neuregulins/metabolism , Rats , Receptor, ErbB-4/genetics , Receptor, ErbB-4/metabolism
2.
J Neurosci ; 37(21): 5232-5249, 2017 05 24.
Article in English | MEDLINE | ID: mdl-28432142

ABSTRACT

The Neuregulin (NRG) family of ErbB ligands is comprised of numerous variants originating from the use of different genes, alternative promoters, and splice variants. NRGs have generally been thought to be transported to axons and presynaptic terminals where they signal via ErbB3/4 receptors in paracrine or juxtacrine mode. However, we recently demonstrated that unprocessed pro-NRG2 accumulates on cell bodies and proximal dendrites, and that NMDAR activity is required for shedding of its ectodomain by metalloproteinases. Here we systematically investigated the subcellular distribution and processing of major NRG isoforms in rat hippocampal neurons. We show that NRG1 isotypes I and II, which like NRG2 are single-pass transmembrane proteins with an Ig-like domain, share the same subcellular distribution and ectodomain shedding properties. We furthermore show that NRG3, like CRD-NRG1, is a dual-pass transmembrane protein that harbors a second transmembrane domain near its amino terminus. Both NRG3 and CRD-NRG1 cluster on axons through juxtacrine interactions with ErbB4 present on GABAergic interneurons. Interestingly, although single-pass NRGs accumulate as unprocessed proforms, axonal puncta of CRD-NRG1 and NRG3 are comprised of processed protein. Mutations of CRD-NRG1 and NRG3 that render them resistant to BACE cleavage, as well as BACE inhibition, result in the loss of axonal puncta and in the accumulation of unprocessed proforms in neuronal soma. Together, these results define two groups of NRGs with distinct membrane topologies and fundamentally different targeting and processing properties in central neurons. The implications of this functional diversity for the regulation of neuronal processes by the NRG/ErbB pathway are discussed.SIGNIFICANCE STATEMENT Numerous Neuregulins (NRGs) are generated through the use of different genes, promoters, and alternative splicing, but the functional significance of this evolutionary conserved diversity remains poorly understood. Here we show that NRGs can be categorized by their membrane topologies. Single-pass NRGs, such as NRG1 Types I/II and NRG2, accumulate as unprocessed proforms on cell bodies, and their ectodomains are shed by metalloproteinases in response to NMDA receptor activation. By contrast, dual-pass CRD-NRG1 and NRG3 are constitutively processed by BACE and accumulate on axons where they interact with ErbB4 in juxtacrine mode. These findings reveal a previously unknown functional relationship between membrane topology, protein processing, and subcellular distribution, and suggest that single- and dual-pass NRGs regulate neuronal functions in fundamentally different ways.


Subject(s)
Neuregulin-1/metabolism , Neurons/metabolism , Signal Transduction , Animals , Aspartic Acid Endopeptidases/metabolism , Axonal Transport , Cells, Cultured , Cerebral Cortex/cytology , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Neuregulin-1/genetics , Neurons/classification , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport , Proteolysis , Rats , Rats, Sprague-Dawley , Receptor, ErbB-4/metabolism
3.
Nat Commun ; 6: 7222, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-26027736

ABSTRACT

The neuregulin receptor ErbB4 is an important modulator of GABAergic interneurons and neural network synchronization. However, little is known about the endogenous ligands that engage ErbB4, the neural processes that activate them or their direct downstream targets. Here we demonstrate, in cultured neurons and in acute slices, that the NMDA receptor is both effector and target of neuregulin 2 (NRG2)/ErbB4 signalling in cortical interneurons. Interneurons co-express ErbB4 and NRG2, and pro-NRG2 accumulates on cell bodies atop subsurface cisternae. NMDA receptor activation rapidly triggers shedding of the signalling-competent NRG2 extracellular domain. In turn, NRG2 promotes ErbB4 association with GluN2B-containing NMDA receptors, followed by rapid internalization of surface receptors and potent downregulation of NMDA but not AMPA receptor currents. These effects occur selectively in ErbB4-positive interneurons and not in ErbB4-negative pyramidal neurons. Our findings reveal an intimate reciprocal relationship between ErbB4 and NMDA receptors with possible implications for the modulation of cortical microcircuits associated with cognitive deficits in psychiatric disorders.


Subject(s)
Feedback, Physiological , GABAergic Neurons/metabolism , Interneurons/metabolism , Nerve Growth Factors/metabolism , Prefrontal Cortex/metabolism , Receptor, ErbB-4/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Blotting, Western , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Fluorescent Antibody Technique , GABAergic Neurons/cytology , HEK293 Cells , Hippocampus/cytology , Humans , Immunohistochemistry , Interneurons/cytology , Mass Spectrometry , Mice , Neuregulin-1 , Neurons , Patch-Clamp Techniques , Prefrontal Cortex/cytology , Rats , Signal Transduction
4.
BMC Dev Biol ; 14: 24, 2014 May 28.
Article in English | MEDLINE | ID: mdl-24886590

ABSTRACT

BACKGROUND: Chloride Intracellular Channel 4 (CLIC4) is one of seven members in the closely related CLIC protein family. CLIC4 is involved in multiple cellular processes including apoptosis, cellular differentiation, inflammation and endothelial tubulogenesis. Despite over a decade of research, no comprehensive in situ expression analysis of CLIC4 in a living organism has been reported. In order to fulfill this goal, we generated a knock-in mouse to express Green Fluorescent Protein (GFP) from the CLIC4 locus, thus substituting the GFP coding region for CLIC4. We used GFP protein expression to eliminate cross reaction with other CLIC family members. RESULTS: We analyzed CLIC4 expression during embryonic development and adult organs. During mid and late gestation, CLIC4 expression is modulated particularly in fetal brain, heart, thymus, liver and kidney as well as in developing brown adipose tissue and stratifying epidermis. In the adult mouse, CLIC4 is highly expressed globally in vascular endothelial cells as well as in liver, lung alveolar septae, pancreatic acini, spermatogonia, renal proximal tubules, cardiomyocytes and thymic epithelial cells. Neural expression included axonal tracks, olfactory bulb, Purkinje cell layer and dentate gyrus. Renal CLIC4 expression was most pronounced in proximal tubules, although altered renal function was not detected in the absence of CLIC4. Myeloid cells and B cells of the spleen are rich in CLIC4 expression as are CD4 and CD8 positive T cells. CONCLUSIONS: In a comprehensive study detailing CLIC4 expression in situ in a mouse model that excludes cross reaction with other family members, we were able to document previously unreported expression for CLIC4 in developing fetus, particularly the brain. In addition, compartmentalized expression of CLIC4 in specific adult tissues and cells provides a focus to explore potential functions of this protein not addressed previously.


Subject(s)
Chloride Channels/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Mitochondrial Proteins/genetics , Adipose Tissue, Brown/embryology , Adipose Tissue, Brown/growth & development , Adipose Tissue, Brown/metabolism , Animals , Brain/embryology , Brain/growth & development , Brain/metabolism , Chloride Channels/metabolism , Epidermis/embryology , Epidermis/growth & development , Epidermis/metabolism , Fetal Heart/embryology , Fetal Heart/metabolism , Green Fluorescent Proteins/metabolism , Heart/growth & development , Immunoblotting , Immunohistochemistry , In Situ Hybridization , Kidney/embryology , Kidney/growth & development , Kidney/metabolism , Liver/embryology , Liver/growth & development , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Mitochondrial Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thymus Gland/embryology , Thymus Gland/growth & development , Thymus Gland/metabolism
5.
Proc Natl Acad Sci U S A ; 110(48): 19603-8, 2013 Nov 26.
Article in English | MEDLINE | ID: mdl-24218551

ABSTRACT

ErbB4 signaling in the central nervous system is implicated in neuropsychiatric disorders and epilepsy. In cortical tissue, ErbB4 associates with excitatory synapses located on inhibitory interneurons. However, biochemical and histological data described herein demonstrate that the vast majority of ErbB4 is extrasynaptic and detergent-soluble. To explore the function of this receptor population, we used unbiased proteomics, in combination with electrophysiological, biochemical, and cell biological techniques, to identify a clinically relevant ErbB4-interacting protein, the GABAA receptor α1 subunit (GABAR α1). We show that ErbB4 and GABAR α1 are robustly coexpressed in hippocampal interneurons, and that ErbB4-null mice have diminished cortical GABAR α1 expression. Moreover, we characterize a Neuregulin-mediated ErbB4 signaling modality, independent of receptor tyrosine kinase activity, that couples ErbB4 to decreased postsynaptic GABAR currents on inhibitory interneurons. Consistent with an evolving understanding of GABAR trafficking, this pathway requires both clathrin-mediated endocytosis and protein kinase C to reduce GABAR inhibitory currents, surface GABAR α1 expression, and colocalization with the inhibitory postsynaptic protein gephyrin. Our results reveal a function of ErbB4, independent of its tyrosine kinase activity, that modulates postsynaptic inhibitory control of hippocampal interneurons and may provide a novel pharmacological target in the treatment of neuropsychiatric disorders and epilepsy.


Subject(s)
ErbB Receptors/metabolism , Hippocampus/cytology , Interneurons/metabolism , Neuregulins/metabolism , Receptors, GABA-A/metabolism , Signal Transduction/physiology , Synapses/metabolism , Animals , Hippocampus/metabolism , Immunohistochemistry , Immunoprecipitation , Mass Spectrometry , Microscopy, Confocal , Patch-Clamp Techniques , Proteomics , Rats , Rats, Sprague-Dawley , Receptor, ErbB-4
6.
Proc Natl Acad Sci U S A ; 109(32): 13118-23, 2012 Aug 07.
Article in English | MEDLINE | ID: mdl-22822214

ABSTRACT

The neuregulin/ErbB signaling network is genetically associated with schizophrenia and modulates hippocampal γ oscillations--a type of neuronal network activity important for higher brain processes and altered in psychiatric disorders. Because neuregulin-1 (NRG-1) dramatically increases extracellular dopamine levels in the hippocampus, we investigated the relationship between NRG/ErbB and dopamine signaling in hippocampal γ oscillations. Using agonists for different D1- and D2-type dopamine receptors, we found that the D4 receptor (D4R) agonist PD168077, but not D1/D5 and D2/D3 agonists, increases γ oscillation power, and its effect is blocked by the highly specific D4R antagonist L-745,870. Using double in situ hybridization and immunofluorescence histochemistry, we show that hippocampal D4R mRNA and protein are more highly expressed in GAD67-positive GABAergic interneurons, many of which express the NRG-1 receptor ErbB4. Importantly, D4 and ErbB4 receptors are coexpressed in parvalbumin-positive basket cells that are critical for γ oscillations. Last, we report that D4R activation is essential for the effects of NRG-1 on network activity because L-745,870 and the atypical antipsychotic clozapine dramatically reduce the NRG-1-induced increase in γ oscillation power. This unique link between D4R and ErbB4 signaling on γ oscillation power, and their coexpression in parvalbumin-expressing interneurons, suggests a cellular mechanism that may be compromised in different psychiatric disorders affecting cognitive control. These findings are important given the association of a DRD4 polymorphism with alterations in attention, working memory, and γ oscillations, and suggest potential benefits of D4R modulators for targeting cognitive deficits.


Subject(s)
Brain Waves/physiology , Dopamine/metabolism , Hippocampus/physiology , Neuregulins/metabolism , Receptors, Dopamine D4/metabolism , Signal Transduction/physiology , Animals , Dopamine/pharmacology , Fluorescent Antibody Technique , Fourier Analysis , Hippocampus/drug effects , Immunohistochemistry , In Situ Hybridization , Interneurons/metabolism , Neuregulins/pharmacology , Pyridines/pharmacology , Pyrroles/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D4/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction
7.
J Neurosci ; 32(9): 2988-97, 2012 Feb 29.
Article in English | MEDLINE | ID: mdl-22378872

ABSTRACT

Neuregulin 1 (NRG-1) and its receptor ErbB4 have emerged as biologically plausible schizophrenia risk factors, modulators of GABAergic and dopaminergic neurotransmission, and as potent regulators of glutamatergic synaptic plasticity. NRG-1 acutely depotentiates LTP in hippocampal slices, and blocking ErbB kinase activity inhibits LTP reversal by theta-pulse stimuli (TPS), an activity-dependent reversal paradigm. NRG-1/ErbB4 signaling in parvalbumin (PV) interneurons has been implicated in inhibitory transmission onto pyramidal neurons. However, the role of ErbB4, in particular in PV interneurons, for LTP reversal has not been investigated. Here we show that ErbB4-null (ErbB4(-/-)) and PV interneuron-restricted mutant (PV-Cre;ErbB4) mice, as well as NRG-1 hypomorphic mice, exhibit increased hippocampal LTP. Moreover, both ErbB4(-/-) and PV-Cre;ErbB4 mice lack TPS-mediated LTP reversal. A comparative behavioral analysis of full and conditional ErbB4 mutant mice revealed that both exhibit hyperactivity in a novel environment and deficits in prepulse inhibition of the startle response. Strikingly, however, only ErbB4(-/-) mice exhibit reduced anxiety-like behaviors in the elevated plus maze task and deficits in cued and contextual fear conditioning. These results suggest that aberrant NRG-1/ErbB4 signaling in PV interneurons accounts for some but not all behavioral abnormalities observed in ErbB4(-/-) mice. Consistent with the observation that PV-Cre;ErbB4 mice exhibit normal fear conditioning, we find that ErbB4 is broadly expressed in the amygdala, largely by cells negative for PV. These findings are important to better understand ErbB4's role in complex behaviors and warrant further analysis of ErbB4 mutant mice lacking the receptor in distinct neuron types.


Subject(s)
ErbB Receptors/physiology , Mental Disorders/metabolism , Neuregulin-1/physiology , Neuronal Plasticity/physiology , Signal Transduction/physiology , Synapses/metabolism , Animals , Fear/physiology , Fear/psychology , Hippocampus/physiology , Long-Term Potentiation/physiology , Male , Maze Learning/physiology , Mental Disorders/psychology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neural Inhibition/physiology , Organ Culture Techniques , Parvalbumins/physiology , Receptor, ErbB-4 , Reflex, Startle/physiology
8.
J Neurosci ; 29(39): 12255-64, 2009 Sep 30.
Article in English | MEDLINE | ID: mdl-19793984

ABSTRACT

NRG1 and ERBB4 have emerged as some of the most reproducible schizophrenia risk genes. Moreover, the Neuregulin (NRG)/ErbB4 signaling pathway has been implicated in dendritic spine morphogenesis, glutamatergic synaptic plasticity, and neural network control. However, despite much attention this pathway and its effects on pyramidal cells have received recently, the presence of ErbB4 in these cells is still controversial. As knowledge of the precise locus of receptor expression is crucial to delineating the mechanisms by which NRG signaling elicits its diverse physiological effects, we have undertaken a thorough analysis of ErbB4 distribution in the CA1 area of the rodent hippocampus using newly generated rabbit monoclonal antibodies and ErbB4-mutant mice as negative controls. We detected ErbB4 immunoreactivity in GABAergic interneurons but not in pyramidal neurons, a finding that was further corroborated by the lack of ErbB4 mRNA in electrophysiologically identified pyramidal neurons as determined by single-cell reverse transcription-PCR. Contrary to some previous reports, we also did not detect processed ErbB4 fragments or nuclear ErbB4 immunoreactivity. Ultrastructural analysis in CA1 interneurons using immunoelectron microscopy revealed abundant ErbB4 expression in the somatodendritic compartment in which it accumulates at, and adjacent to, glutamatergic postsynaptic sites. In contrast, we found no evidence for presynaptic expression in cultured GAD67-positive hippocampal interneurons and in CA1 basket cell terminals. Our findings identify ErbB4-expressing interneurons, but not pyramidal neurons, as a primary target of NRG signaling in the hippocampus and, furthermore, implicate ErbB4 as a selective marker for glutamatergic synapses on inhibitory interneurons.


Subject(s)
ErbB Receptors/biosynthesis , Gene Expression Regulation, Enzymologic , Hippocampus/enzymology , Interneurons/enzymology , Pyramidal Cells/enzymology , Animals , Cells, Cultured , ErbB Receptors/deficiency , ErbB Receptors/genetics , Hippocampus/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Pyramidal Cells/ultrastructure , Rabbits , Rats , Rats, Sprague-Dawley , Receptor, ErbB-4 , Species Specificity , gamma-Aminobutyric Acid/physiology
9.
Development ; 134(21): 3827-36, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17913785

ABSTRACT

Specification of cell lineages in mammals begins shortly after fertilization with formation of a blastocyst consisting of trophectoderm, which contributes exclusively to the placenta, and inner cell mass (ICM), from which the embryo develops. Here we report that ablation of the mouse Tead4 gene results in a preimplantation lethal phenotype, and TEAD4 is one of two highly homologous TEAD transcription factors that are expressed during zygotic gene activation in mouse 2-cell embryos. Tead4(-/-) embryos do not express trophectoderm-specific genes, such as Cdx2, but do express ICM-specific genes, such as Oct4 (also known as Pou5f1). Consequently, Tead4(-/-) morulae do not produce trophoblast stem cells, trophectoderm or blastocoel cavities, and therefore do not implant into the uterine endometrium. However, Tead4(-/-) embryos can produce embryonic stem cells, a derivative of ICM, and if the Tead4 allele is not disrupted until after implantation, then Tead4(-/-) embryos complete development. Thus, Tead4 is the earliest gene shown to be uniquely required for specification of the trophectoderm lineage.


Subject(s)
Cell Lineage , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Muscle Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Embryo Implantation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Genotype , Male , Mice , Mice, Knockout , Muscle Proteins/deficiency , Muscle Proteins/genetics , TEA Domain Transcription Factors , Transcription Factors/deficiency , Transcription Factors/genetics , Transcriptional Activation
10.
Mol Cell Neurosci ; 34(3): 468-80, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17276696

ABSTRACT

NMDA receptor "knock-in" mice were generated by inserting the nuclear beta-galactosidase reporter at the NR2C subunit translation initiation site. Novel cell types and dynamic patterns of NR2C expression were identified using these mice, which were unnoticed before because reagents that specifically recognize NR2C-containing receptors are non-existent. We identified a transition zone from NR2C-expressing neurons to astrocytes in an area connecting the retrosplenial cortex and hippocampus. We demonstrate that NR2C is expressed in a subset of S100beta-positive/GFAP-negative glial cells in the striatum, olfactory bulb and cerebral cortex. We also demonstrate novel areas of neuronal expression such as retrosplenial cortex, thalamus, pontine and vestibular nuclei. In addition, we show that during cerebellar development NR2C is expressed in transient caudal-rostral gradients and parasagittal bands in subsets of granule cells residing in the internal granular layer, further demonstrating heterogeneity of granule neurons. These results point to novel functions of NR2C-containing NMDA receptors.


Subject(s)
Brain/metabolism , Gene Expression Regulation, Developmental/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , beta-Galactosidase/metabolism , Animals , Animals, Newborn , Brain/cytology , Brain/growth & development , Immunohistochemistry/methods , In Situ Hybridization , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , beta-Galactosidase/genetics
11.
J Comp Neurol ; 472(2): 156-72, 2004 Apr 26.
Article in English | MEDLINE | ID: mdl-15048684

ABSTRACT

Neuregulin-1 (NRG-1) regulates numerous aspects of neural development and synaptic plasticity; the functions of NRG-2 and NRG-3 are presently unknown. As a first step toward understanding how NRGs contribute to distinct aspects of neural development and function, we characterized their regional and subcellular expression patterns in developing brain. The expression of NRG-1-3 mRNAs was compared postnatally (P0, P7, adult) by using in situ hybridization. NRG-1 expression is highest at birth, whereas NRG-2 mRNA levels increase with development; expression of both genes is restricted to distinct brain regions. In contrast, NRG-3 transcripts are abundant in most brain regions throughout development. NRG-2 antibodies were generated to analyze protein processing, expression, and subcellular distribution. As with NRG-1, the transmembrane NRG-2 proprotein is proteolytically processed in transfected HEK 293 cells and in neural tissues, and its ectodomain is exposed and accumulates on the neuron surface. Despite the structural similarities between NRG-1 and NRG-2, we unexpectedly found that NRG-2 colocalizes with MAP2 in proximal primary dendrites of hippocampal neurons in culture and in vivo, although it is not detectable in axons or in axon terminals. These findings were confirmed with NRG-2 ectodomain antisera and epitope-tagged recombinant protein. In cerebellum, NRG-2 colocalizes with calbindin in proximal dendrites and soma of Purkinje cells. In contrast, NRG-1 is highly expressed in axons of dissociated hippocampal neurons, as well as in somas and dendrites. The distinct temporal, regional, and subcellular expression of NRG-2 suggests its unique and nonredundant role in neural function.


Subject(s)
Dendrites/metabolism , Gene Expression Regulation, Developmental/physiology , Hippocampus/growth & development , Nerve Growth Factors/metabolism , Animals , Animals, Newborn , Cells, Cultured , Dendrites/chemistry , Dendrites/genetics , Hippocampus/cytology , Hippocampus/physiology , Humans , Mice , Nerve Growth Factors/biosynthesis , Nerve Growth Factors/genetics , Neuregulin-1/metabolism , Neurons/chemistry , Neurons/metabolism , Protein Processing, Post-Translational , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Transfection
12.
Hum Mol Genet ; 11(11): 1291-301, 2002 May 15.
Article in English | MEDLINE | ID: mdl-12019210

ABSTRACT

Mutations in the MYOC gene may lead to juvenile open-angle glaucoma with high intraocular pressure, and are detected in about 4% of people with adult onset glaucoma. Most of these mutations are found in the third exon of the gene encoding the olfactomedin-like domain located at the C terminus of the protein. Another olfactomedin-related protein, known as noelin or pancortin, is involved in the generation of neural crest cells. Here we describe the identification of a novel olfactomedin-related gene, named optimedin, located on chromosome 1p21 in humans. Optimedin and noelin are both expressed in brain and retina. However, unlike noelin, rat optimedin is also highly expressed in the epithelial cells of the iris and the ciliary body in close proximity to the sites of Myoc expression. In the human eye, optimedin is expressed in the retina and the trabecular meshwork. Both optimedin and myocilin are localized in Golgi and are secreted proteins. The presence of mutant myocilin interferes with secretion of optimedin in transfected cells. Optimedin and myocilin interact with each other in vitro as judged by the GST pulldown, co-immunoprecipitation and far-western binding assays. The C-terminal olfactomedin domains are essential for interaction between optimedin and myocilin, while the N-terminal domains of both proteins are involved in the formation of protein homodimers. We suggest that optimedin may be a candidate gene for disorders involving the anterior segment of the eye and the retina.


Subject(s)
Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Eye Proteins/metabolism , Eye/metabolism , Glaucoma/genetics , Glycoproteins/genetics , Glycoproteins/metabolism , Amino Acid Sequence , Animals , Blotting, Northern , COS Cells , Chlorocebus aethiops , Cytoskeletal Proteins , Extracellular Matrix Proteins/isolation & purification , Eye Proteins/genetics , Gene Expression , Glaucoma/metabolism , Glycoproteins/isolation & purification , Humans , In Situ Hybridization , Molecular Sequence Data , Organ Specificity , Rats , Sequence Alignment
13.
Mol Cell Neurosci ; 19(1): 88-96, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11817900

ABSTRACT

In ventromedial cells of the developing CNS, Sonic hedgehog (Shh) has been shown to affect precursor proliferation, phenotype determination, and survival. Here we show that Shh and its receptor, Ptc-1, are expressed in the adult rat basal forebrain, and that Ptc-1 is expressed specifically by cholinergic neurons. In basal forebrain cultures, Shh was added alone and in combination with nerve growth factor (NGF), and the number of cholinergic neurons was determined by choline acetyltransferase (ChAT) immunocytochemistry. By 8 days in vitro, Shh and NGF show a synergistic effect: the number of ChAT-positive cells after treatment with both factors is increased over untreated cultures or cultures treated with either factor alone. While Shh increases the overall basal level of proliferation, double-labeling of dividing neuronal precursors with [(3)H]thymidine followed by ChAT immunocytochemistry after they mature, demonstrates that the specific increase in cholinergic neurons is not due to this proliferation enhancement. These experiments imply a role for Shh in the development of postmitotic cholinergic neurons and suggest a therapeutic value for Shh in neurodegenerative disease.


Subject(s)
Nerve Growth Factor/pharmacology , Neurons/cytology , Neurons/drug effects , Trans-Activators/pharmacology , Acetylcholine/physiology , Age Factors , Alzheimer Disease/drug therapy , Animals , Cell Count , Cell Division/drug effects , Cells, Cultured , Choline O-Acetyltransferase/analysis , Drug Synergism , Gene Expression/physiology , Hedgehog Proteins , Membrane Proteins/analysis , Membrane Proteins/genetics , Mice , Mice, Transgenic , Neurons/enzymology , Patched Receptors , Patched-1 Receptor , Prosencephalon/cytology , RNA, Messenger/analysis , Receptors, Cell Surface , Trans-Activators/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...