Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cell Rep ; 42(9): 113068, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37656624

ABSTRACT

Primary somatosensory axons stop regenerating as they re-enter the spinal cord, resulting in incurable sensory loss. What arrests them has remained unclear. We previously showed that axons stop by forming synaptic contacts with unknown non-neuronal cells. Here, we identified these cells in adult mice as oligodendrocyte precursor cells (OPCs). We also found that only a few axons stop regenerating by forming dystrophic endings, exclusively at the CNS:peripheral nervous system (PNS) borderline where OPCs are absent. Most axons stop in contact with a dense network of OPC processes. Live imaging, immuno-electron microscopy (immuno-EM), and OPC-dorsal root ganglia (DRG) co-culture additionally suggest that axons are rapidly immobilized by forming synapses with OPCs. Genetic OPC ablation enables many axons to continue regenerating deep into the spinal cord. We propose that sensory axons stop regenerating by encountering OPCs that induce presynaptic differentiation. Our findings identify OPCs as a major regenerative barrier that prevents intraspinal restoration of sensory circuits following spinal root injury.


Subject(s)
Oligodendrocyte Precursor Cells , Mice , Animals , Spinal Cord/physiology , Axons/physiology , Spinal Nerve Roots , Ganglia, Spinal/physiology , Nerve Regeneration/physiology
2.
Front Cell Neurosci ; 16: 847097, 2022.
Article in English | MEDLINE | ID: mdl-35465615

ABSTRACT

Oligodendrocytes (OLs) form myelin sheaths and provide metabolic support to axons in the CNS. Although most OLs develop during early postnatal life, OL generation continues in adulthood, and this late oligodendrogenesis may contribute to neuronal network plasticity in the adult brain. We used genetic tools for OL labeling and fate tracing of OL progenitors (OPCs), thereby determining OL population growth in hippocampal subregions with normal aging. OL numbers increased up to at least 1 year of age, but the rates and degrees of this OL change differed among hippocampal subregions. In particular, adult oligodendrogenesis was most prominent in the CA3 and CA4 subregions. In Alzheimer's disease-like conditions, OL loss was also most severe in the CA3 and CA4 of APP/PS1 mice, although the disease did not impair the rate of OPC differentiation into OLs in those regions. Such region-specific, dynamic OL changes were not correlated with those of OPCs or astrocytes, or the regional distribution of Aß deposits. Our findings suggest subregion-dependent mechanisms for myelin plasticity and disease-associated OL vulnerability in the adult hippocampus.

3.
Cell Rep ; 35(7): 109147, 2021 05 18.
Article in English | MEDLINE | ID: mdl-34010640

ABSTRACT

Oligodendrocyte precursor cells (OPCs) are essential for developmental myelination and oligodendrocyte regeneration after CNS injury. These progenitors express calcium-permeable AMPA receptors (AMPARs) and form direct synapses with neurons throughout the CNS, but the roles of this signaling are unclear. To enable selective alteration of the properties of AMPARs in oligodendroglia, we generate mice that allow cell-specific overexpression of EGFP-GluA2 in vivo. In healthy conditions, OPC-specific GluA2 overexpression significantly increase their proliferation in an age-dependent manner but did not alter their rate of differentiation into oligodendrocytes. In contrast, after demyelinating brain injury in neonates or adults, higher GluA2 levels promote both OPC proliferation and oligodendrocyte regeneration, but do not prevent injury-induced initial cell loss. These findings indicate that AMPAR GluA2 content regulates the proliferative and regenerative behavior of adult OPCs, serving as a putative target for better myelin repair.


Subject(s)
Oligodendrocyte Precursor Cells/metabolism , Oligodendroglia/metabolism , Animals , Cell Proliferation , Mice , Rats , Receptors, AMPA , Regeneration
4.
Elife ; 72018 02 20.
Article in English | MEDLINE | ID: mdl-29461205

ABSTRACT

Oligodendrocytes (OLs), the myelin-forming CNS glia, are highly vulnerable to cellular stresses, and a severe myelin loss underlies numerous CNS disorders. Expedited OL regeneration may prevent further axonal damage and facilitate functional CNS repair. Although adult OL progenitors (OPCs) are the primary players for OL regeneration, targetable OPC-specific intracellular signaling mechanisms for facilitated OL regeneration remain elusive. Here, we report that OPC-targeted PTEN inactivation in the mouse, in contrast to OL-specific manipulations, markedly promotes OL differentiation and regeneration in the mature CNS. Unexpectedly, an additional deletion of mTOR did not reverse the enhanced OL development from PTEN-deficient OPCs. Instead, ablation of GSK3ß, another downstream signaling molecule that is negatively regulated by PTEN-Akt, enhanced OL development. Our results suggest that PTEN persistently suppresses OL development in an mTOR-independent manner, and at least in part, via controlling GSK3ß activity. OPC-targeted PTEN-GSK3ß inactivation may benefit facilitated OL regeneration and myelin repair.


Subject(s)
Cell Differentiation , Glycogen Synthase Kinase 3 beta/metabolism , Oligodendroglia/physiology , PTEN Phosphohydrolase/metabolism , Signal Transduction , Stem Cells/physiology , TOR Serine-Threonine Kinases/metabolism , Animals , Mice
5.
J Neurosci ; 37(36): 8635-8654, 2017 09 06.
Article in English | MEDLINE | ID: mdl-28760862

ABSTRACT

Spontaneous remyelination occurs after spinal cord injury (SCI), but the extent of myelin repair and identity of the cells responsible remain incompletely understood and contentious. We assessed the cellular origin of new myelin by fate mapping platelet-derived growth factor receptor α (PDGFRα), Olig2+, and P0+ cells following contusion SCI in mice. Oligodendrocyte precursor cells (OPCs; PDGFRα+) produced oligodendrocytes responsible for de novo ensheathment of ∼30% of myelinated spinal axons at injury epicenter 3 months after SCI, demonstrating that these resident cells are a major contributor to oligodendrocyte regeneration. OPCs also produced the majority of myelinating Schwann cells in the injured spinal cord; invasion of peripheral myelinating (P0+) Schwann cells made only a limited contribution. These findings reveal that PDGFRα+ cells perform diverse roles in CNS repair, as multipotential progenitors that generate both classes of myelinating cells. This endogenous repair might be exploited as a therapeutic target for CNS trauma and disease.SIGNIFICANCE STATEMENT Spinal cord injury (SCI) leads to profound functional deficits, though substantial numbers of axons often survive. One possible explanation for these deficits is loss of myelin, creating conduction block at the site of injury. SCI leads to oligodendrocyte death and demyelination, and clinical trials have tested glial transplants to promote myelin repair. However, the degree and duration of myelin loss, and the extent and mechanisms of endogenous repair, have been contentious issues. Here, we use genetic fate mapping to demonstrate that spontaneous myelin repair by endogenous oligodendrocyte precursors is much more robust than previously recognized. These findings are relevant to many types of CNS pathology, raising the possibility that CNS precursors could be manipulated to repair myelin in lieu of glial transplantation.


Subject(s)
Myelin Sheath/pathology , Nerve Regeneration/physiology , Neural Stem Cells/pathology , Neuronal Plasticity , Oligodendroglia/physiology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Animals , Cell Differentiation , Cell Proliferation , Female , Male , Mice
6.
Curr Biol ; 27(12): 1791-1800.e5, 2017 Jun 19.
Article in English | MEDLINE | ID: mdl-28578927

ABSTRACT

The mammalian pupillary light reflex (PLR) involves a bilateral brain circuit whereby afferent light signals in the optic nerve ultimately drive iris-sphincter-muscle contraction via excitatory cholinergic parasympathetic innervation [1, 2]. Additionally, the PLR in nocturnal and crepuscular sub-primate mammals has a "local" component in the isolated sphincter muscle [3-5], as in amphibians, fish, and bird [6-10]. In mouse, this local PLR requires the pigment melanopsin [5], originally found in intrinsically photosensitive retinal ganglion cells (ipRGCs) [11-19]. However, melanopsin's presence and effector pathway locally in the iris remain uncertain. The sphincter muscle itself may express melanopsin [5], or its cholinergic parasympathetic innervation may be modulated by suggested intraocular axonal collaterals of ipRGCs traveling to the eye's ciliary body or even to the iris [20-22]. Here, we show that the muscarinic receptor antagonist, atropine, eliminated the effect of acetylcholine (ACh), but not of light, on isolated mouse sphincter muscle. Conversely, selective genetic deletion of melanopsin in smooth muscle mostly removed the light-induced, but not the ACh-triggered, increase in isolated sphincter muscle's tension and largely suppressed the local PLR in vivo. Thus, sphincter muscle cells are bona fide, albeit unconventional, photoreceptors. We found melanopsin expression in a small subset of mouse iris sphincter muscle cells, with the light-induced contractile signal apparently spreading through gap junctions into neighboring muscle cells. Light and ACh share a common signaling pathway in sphincter muscle. In summary, our experiments have provided details of a photosignaling process in the eye occurring entirely outside the retina.


Subject(s)
Acetylcholine/metabolism , Atropine/pharmacology , Iris/drug effects , Light , Muscarinic Antagonists/pharmacology , Signal Transduction , Animals , Mice , Mice, Transgenic , Photic Stimulation , Signal Transduction/drug effects
7.
Elife ; 62017 01 26.
Article in English | MEDLINE | ID: mdl-28124973

ABSTRACT

Nuclear exclusion of the transcriptional regulators and potent oncoproteins, YAP/TAZ, is considered necessary for adult tissue homeostasis. Here we show that nuclear YAP/TAZ are essential regulators of peripheral nerve development and myelin maintenance. To proliferate, developing Schwann cells (SCs) require YAP/TAZ to enter S-phase and, without them, fail to generate sufficient SCs for timely axon sorting. To differentiate, SCs require YAP/TAZ to upregulate Krox20 and, without them, completely fail to myelinate, resulting in severe peripheral neuropathy. Remarkably, in adulthood, nuclear YAP/TAZ are selectively expressed by myelinating SCs, and conditional ablation results in severe peripheral demyelination and mouse death. YAP/TAZ regulate both developmental and adult myelination by driving TEAD1 to activate Krox20. Therefore, YAP/TAZ are crucial for SCs to myelinate developing nerve and to maintain myelinated nerve in adulthood. Our study also provides a new insight into the role of nuclear YAP/TAZ in homeostatic maintenance of an adult tissue.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Myelin Sheath/metabolism , Phosphoproteins/metabolism , Schwann Cells/physiology , Transcription Factors/metabolism , Acyltransferases , Animals , Cell Cycle Proteins , Cell Differentiation , Cell Proliferation , Mice , YAP-Signaling Proteins
8.
J Neurosci ; 34(49): 16369-84, 2014 Dec 03.
Article in English | MEDLINE | ID: mdl-25471575

ABSTRACT

NG2 is purportedly one of the most growth-inhibitory chondroitin sulfate proteoglycans (CSPGs) produced after spinal cord injury. Nonetheless, once the severed axon tips dieback from the lesion core into the penumbra they closely associate with NG2+ cells. We asked if proteoglycans play a role in this tight cell-cell interaction and whether overadhesion upon these cells might participate in regeneration failure in rodents. Studies using varying ratios of CSPGs and adhesion molecules along with chondroitinase ABC, as well as purified adult cord-derived NG2 glia, demonstrate that CSPGs are involved in entrapping neurons. Once dystrophic axons become stabilized upon NG2+ cells, they form synaptic-like connections both in vitro and in vivo. In NG2 knock-out mice, sensory axons in the dorsal columns dieback further than their control counterparts. When axons are double conditioned to enhance their growth potential, some traverse the lesion core and express reduced amounts of synaptic proteins. Our studies suggest that proteoglycan-mediated entrapment upon NG2+ cells is an additional obstacle to CNS axon regeneration.


Subject(s)
Antigens/physiology , Axons/physiology , Cell Communication/physiology , Nerve Regeneration/physiology , Proteoglycans/physiology , Spinal Cord Injuries/physiopathology , Synapses/physiology , Animals , Antigens/genetics , Axons/ultrastructure , Cell Tracking , Cells, Cultured , Chondroitin Sulfate Proteoglycans/physiology , Fibronectins/physiology , Ganglia, Spinal/physiopathology , Ganglia, Spinal/ultrastructure , Integrin beta1/physiology , Laminin/physiology , Mice , Mice, Knockout , Nerve Degeneration/physiopathology , Proteoglycans/genetics
9.
J Comp Neurol ; 522(6): 1316-32, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24151158

ABSTRACT

After spinal cord transection, lampreys recover functionally and axons regenerate. It is not known whether this is accompanied by neurogenesis. Previous studies suggested a baseline level of nonneuronal cell proliferation in the spinal cord and rhombencephalon (where most supraspinal projecting neurons are located). To determine whether cell proliferation increases after injury and whether this includes neurogenesis, larval lampreys were spinally transected and injected with 5-bromo-2&prime-deoxyuridine (BrdU) at 0-3 weeks posttransection. Labeled cells were counted in the lesion site, within 0.5 mm rostral and caudal to the lesion, and in the rhombencephalon. One group of animals was processed in the winter and a second group was processed in the summer. The number of labeled cells was greater in winter than in summer. The lesion site had the most BrdU labeling at all times, correlating with an increase in the number of cells. In the adjacent spinal cord, the percentage of BrdU labeling was higher in the ependymal than in nonependymal regions. This was also true in the rhombencephalon but only in summer. In winter, BrdU labeling was seen primarily in the subventricular and peripheral zones. Some BrdU-labeled cells were also double labeled by antibodies to glial-specific (antikeratin) as well as neuron-specific (anti-Hu) antigens, indicating that both gliogenesis and neurogenesis occurred after spinal cord transection. However, the new neurons were restricted to the ependymal zone, were never labeled by antineurofilament antibodies, and never migrated away from the ependyma even at 5 weeks after BrdU injection. They would appear to be cerebrospinal fluid-contacting neurons.


Subject(s)
Central Nervous System/physiopathology , Neurogenesis/physiology , Spinal Cord Injuries/pathology , Animals , Bromodeoxyuridine/metabolism , Cell Proliferation , Keratins/metabolism , Lampreys , Nerve Tissue Proteins/metabolism
10.
Nat Neurosci ; 16(6): 668-76, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23624515

ABSTRACT

The adult CNS contains an abundant population of oligodendrocyte precursor cells (NG2(+) cells) that generate oligodendrocytes and repair myelin, but how these ubiquitous progenitors maintain their density is unknown. We generated NG2-mEGFP mice and used in vivo two-photon imaging to study their dynamics in the adult brain. Time-lapse imaging revealed that NG2(+) cells in the cortex were highly dynamic; they surveyed their local environment with motile filopodia, extended growth cones and continuously migrated. They maintained unique territories though self-avoidance, and NG2(+) cell loss though death, differentiation or ablation triggered rapid migration and proliferation of adjacent cells to restore their density. NG2(+) cells recruited to sites of focal CNS injury were similarly replaced by a proliferative burst surrounding the injury site. Thus, homeostatic control of NG2(+) cell density through a balance of active growth and self-repulsion ensures that these progenitors are available to replace oligodendrocytes and participate in tissue repair.


Subject(s)
Cell Differentiation/physiology , Cerebral Cortex/cytology , Homeostasis/physiology , Neural Stem Cells/physiology , Oligodendroglia/physiology , Animals , Cell Count , Cell Death/physiology , Cell Movement/physiology , Cell Proliferation , Cerebral Cortex/physiology , Mice , Mice, Transgenic , Time-Lapse Imaging
11.
Nat Neurosci ; 16(5): 571-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23542689

ABSTRACT

Oligodendrocytes associate with axons to establish myelin and provide metabolic support to neurons. In the spinal cord of amyotrophic lateral sclerosis (ALS) mice, oligodendrocytes downregulate transporters that transfer glycolytic substrates to neurons and oligodendrocyte progenitors (NG2(+) cells) exhibit enhanced proliferation and differentiation, although the cause of these changes in oligodendroglia is unknown. We found extensive degeneration of gray matter oligodendrocytes in the spinal cord of SOD1 (G93A) ALS mice prior to disease onset. Although new oligodendrocytes were formed, they failed to mature, resulting in progressive demyelination. Oligodendrocyte dysfunction was also prevalent in human ALS, as gray matter demyelination and reactive changes in NG2(+) cells were observed in motor cortex and spinal cord of ALS patients. Selective removal of mutant SOD1 from oligodendroglia substantially delayed disease onset and prolonged survival in ALS mice, suggesting that ALS-linked genes enhance the vulnerability of motor neurons and accelerate disease by directly impairing the function of oligodendrocytes.


Subject(s)
Amyotrophic Lateral Sclerosis/complications , Amyotrophic Lateral Sclerosis/pathology , Nerve Degeneration/etiology , Oligodendroglia/pathology , Regeneration/physiology , Animals , Animals, Newborn , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Functional Laterality/genetics , Humans , Mice , Mice, Transgenic , Motor Cortex/pathology , Myelin Sheath/metabolism , Myelin Sheath/pathology , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Nerve Fibers, Myelinated/pathology , Nerve Fibers, Myelinated/ultrastructure , Nerve Tissue Proteins/metabolism , Oligodendroglia/drug effects , Oligodendroglia/ultrastructure , Receptor, Platelet-Derived Growth Factor alpha/genetics , Regeneration/drug effects , Regeneration/genetics , Spinal Cord/cytology , Spinal Cord/pathology , Survival Analysis
12.
J Neurosci ; 31(35): 12650-62, 2011 Aug 31.
Article in English | MEDLINE | ID: mdl-21880926

ABSTRACT

Oligodendrocyte precursor cells (OPCs) express NMDA receptors (NMDARs) and form synapses with glutamatergic neurons throughout the CNS. Although glutamate influences the proliferation and maturation of these progenitors in vitro, the role of NMDAR signaling in oligodendrogenesis and myelination in vivo is not known. Here, we investigated the consequences of genetically deleting the obligatory NMDAR subunit NR1 from OPCs and their oligodendrocyte progeny in the CNS of developing and mature mice. NMDAR-deficient OPCs proliferated normally, achieved appropriate densities in gray and white matter, and differentiated to form major white matter tracts without delay. OPCs also retained their characteristic physiological and morphological properties in the absence of NMDAR signaling and were able to form synapses with glutamatergic axons. However, expression of calcium-permeable AMPA receptors (AMPARs) was enhanced in NMDAR-deficient OPCs. These results suggest that NMDAR signaling is not used to control OPC development but to regulate AMPAR-dependent signaling with surrounding axons, pointing to additional functions for these ubiquitous glial cells.


Subject(s)
Brain/cytology , Cell Proliferation , Oligodendroglia/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction/physiology , Stem Cells/physiology , Age Factors , Aldehyde Dehydrogenase 1 Family , Analysis of Variance , Animals , Animals, Newborn , Autophagy-Related Proteins , Basic Helix-Loop-Helix Transcription Factors/genetics , Biophysics , Brain/growth & development , Bromodeoxyuridine/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Gene Expression Regulation, Developmental/genetics , Glial Fibrillary Acidic Protein/metabolism , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/metabolism , Isoenzymes/metabolism , Luminescent Proteins/genetics , Mice , Mice, Transgenic , Myelin Basic Protein/metabolism , Patch-Clamp Techniques , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Receptors, N-Methyl-D-Aspartate/deficiency , Retinal Dehydrogenase/metabolism , Signal Transduction/genetics , Synapses/genetics , Synapses/physiology
13.
Neuron ; 68(4): 668-81, 2010 Nov 18.
Article in English | MEDLINE | ID: mdl-21092857

ABSTRACT

The mammalian CNS contains a ubiquitous population of glial progenitors known as NG2+ cells that have the ability to develop into oligodendrocytes and undergo dramatic changes in response to injury and demyelination. Although it has been reported that NG2+ cells are multipotent, their fate in health and disease remains controversial. Here, we generated PDGFαR-CreER transgenic mice and followed their fate in vivo in the developing and adult CNS. These studies revealed that NG2+ cells in the postnatal CNS generate myelinating oligodendrocytes, but not astrocytes or neurons. In regions of neurodegeneration in the spinal cord of ALS mice, NG2+ cells exhibited enhanced proliferation and accelerated differentiation into oligodendrocytes but remained committed to the oligodendrocyte lineage. These results indicate that NG2+ cells in the normal CNS are oligodendrocyte precursors with restricted lineage potential and that cell loss and gliosis are not sufficient to alter the lineage potential of these progenitors.


Subject(s)
Cell Lineage , Nerve Degeneration/pathology , Neuroglia/cytology , Oligodendroglia/cytology , Stem Cells/cytology , Animals , Animals, Newborn , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Proliferation , Disease Models, Animal , Mice , Mice, Transgenic , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Neurogenesis/genetics , Neuroglia/metabolism , Neuroglia/ultrastructure , Oligodendroglia/pathology , Oligodendroglia/ultrastructure , Random Allocation , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/physiology , Spinal Cord/cytology , Spinal Cord/metabolism , Spinal Cord/ultrastructure , Stem Cells/pathology , Stem Cells/ultrastructure
14.
Nature ; 457(7227): 281-7, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19118382

ABSTRACT

A subset of retinal ganglion cells has recently been discovered to be intrinsically photosensitive, with melanopsin as the pigment. These cells project primarily to brain centres for non-image-forming visual functions such as the pupillary light reflex and circadian photoentrainment. How well they signal intrinsic light absorption to drive behaviour remains unclear. Here we report fundamental parameters governing their intrinsic light responses and associated spike generation. The membrane density of melanopsin is 10(4)-fold lower than that of rod and cone pigments, resulting in a very low photon catch and a phototransducing role only in relatively bright light. Nonetheless, each captured photon elicits a large and extraordinarily prolonged response, with a unique shape among known photoreceptors. Notably, like rods, these cells are capable of signalling single-photon absorption. A flash causing a few hundred isomerized melanopsin molecules in a retina is sufficient for reaching threshold for the pupillary light reflex.


Subject(s)
Photons , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/radiation effects , Rod Opsins/metabolism , Action Potentials/radiation effects , Animals , Brain/metabolism , Kinetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pupil/physiology , Pupil/radiation effects , Reflex, Pupillary/radiation effects
16.
Neuron ; 43(3): 401-13, 2004 Aug 05.
Article in English | MEDLINE | ID: mdl-15294147

ABSTRACT

Drug addiction involves complex interactions between pharmacology and learning in genetically susceptible individuals. Members of the Homer gene family are regulated by acute and chronic cocaine administration. Here, we report that deletion of Homer1 or Homer2 in mice caused the same increase in sensitivity to cocaine-induced locomotion, conditioned reward, and augmented extracellular glutamate in nucleus accumbens as that elicited by withdrawal from repeated cocaine administration. Moreover, adeno-associated virus-mediated restoration of Homer2 in the accumbens of Homer2 KO mice reversed the cocaine-sensitized phenotype. Further analysis of Homer2 KO mice revealed extensive additional behavioral and neurochemical similarities to cocaine-sensitized animals, including accelerated acquisition of cocaine self-administration and altered regulation of glutamate by metabotropic glutamate receptors and cystine/glutamate exchange. These data show that Homer deletion mimics the behavioral and neurochemical phenotype produced by repeated cocaine administration and implicate Homer in regulating addiction to cocaine.


Subject(s)
Carrier Proteins/physiology , Cocaine-Related Disorders/metabolism , Cocaine/administration & dosage , Animals , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cocaine-Related Disorders/genetics , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Dose-Response Relationship, Drug , Glutamic Acid/metabolism , Homer Scaffolding Proteins , Locomotion/drug effects , Locomotion/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Self Administration
17.
Cell ; 114(6): 777-89, 2003 Sep 19.
Article in English | MEDLINE | ID: mdl-14505576

ABSTRACT

Receptor signaling at the plasma membrane often releases calcium from intracellular stores. For example, inositol triphosphate (IP3) produced by receptor-coupled phospholipase C activates an intracellular store calcium channel, the IP(3)R. Conversely, stores can induce extracellular calcium to enter the cell through plasma membrane channels, too. How this "reverse" coupling works was unclear, but store IP(3)Rs were proposed to bind and regulate plasma membrane TRP cation channels. Here, we demonstrate that the adaptor protein, termed Homer, facilitates a physical association between TRPC1 and the IP(3)R that is required for the TRP channel to respond to signals. The TRPC1-Homer-IP(3)R complex is dynamic and its disassembly parallels TRPC1 channel activation. Homer's action depends on its ability to crosslink and is blocked by the dominant-negative immediate early gene form, H1a. Since H1a is transcriptionally regulated by cellular activity, this mechanism can affect both short and long-term regulation of TRPC1 function.


Subject(s)
Calcium Channels/metabolism , Carrier Proteins/metabolism , Ion Channel Gating/physiology , Neuropeptides/metabolism , Receptors, Cell Surface/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Binding Sites/physiology , Brain/metabolism , Calcium/metabolism , Calcium Channels/genetics , Calcium Signaling/genetics , Cell Membrane/metabolism , Cells, Cultured , Homer Scaffolding Proteins , Inositol 1,4,5-Trisphosphate Receptors , Macromolecular Substances , Membrane Potentials/genetics , Mice , Mice, Mutant Strains , Mutation/genetics , Patch-Clamp Techniques , Protein Binding/genetics , Protein Structure, Tertiary/physiology , Rats , TRPC Cation Channels
18.
J Neurosci ; 22(1): 167-75, 2002 Jan 01.
Article in English | MEDLINE | ID: mdl-11756499

ABSTRACT

Three Homer genes regulate the activity of metabotropic glutamate receptors mGluR1a and mGluR5 and their coupling to releasable intracellular Ca2+ pools and ion channels. Only the Homer 1 gene evolved bimodal expression of constitutive (Homer 1b and c) and immediate early gene (IEG) products (Homer 1a and Ania 3). The IEG forms compete functionally with the constitutive Homer proteins. The complex expression of the Homer 1 gene, unique for IEGs, focused our attention on the gene organization. In contrast to most IEGs, which have genes that are <5 kb, the Homer 1 gene was found to span approximately 100 kb. The constitutive Homer 1b/c forms are encoded by exons 1-10, whereas the IEG forms are encoded by exons 1-5 and parts of intron 5. RNase protection demonstrated a >10-fold activity-dependent increase in mRNA levels exclusively for the IEG forms. Moreover, fluorescent in situ hybridization documented that new primary Homer 1 transcripts are induced in neuronal nuclei within a few minutes after seizure, typical of IEGs, and that Homer 1b-specific exons are excluded from the activity-induced transcripts. Thus, at the resting state of the neurons, the entire gene is constitutively transcribed at low levels to yield Homer 1b/c transcripts. Neuronal activity sharply increases the rate of transcription initiation, with most transcripts now ending within the central intron. These coordinate transcriptional events rapidly convert a constitutive gene to an IEG and regulate the expression of functionally different Homer 1 proteins.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Gene Expression Regulation/physiology , Genes, Immediate-Early/physiology , Neuropeptides/genetics , Neuropeptides/metabolism , Synapses/metabolism , Alternative Splicing , Animals , DNA, Complementary/genetics , Exons/physiology , Genomic Library , Homer Scaffolding Proteins , In Situ Hybridization , Introns/physiology , Mice , Molecular Sequence Data , Neurons/cytology , Neurons/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Analysis, DNA , Synaptic Transmission/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...