Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Neurosci ; 32(21): 7301-10, 2012 May 23.
Article in English | MEDLINE | ID: mdl-22623675

ABSTRACT

G-protein-coupled receptors (GPCRs) mediate numerous physiological functions and represent prime therapeutic targets. Receptor trafficking upon agonist stimulation is critical for GPCR function, but examining this process in vivo remains a true challenge. Using knock-in mice expressing functional fluorescent delta opioid receptors under the control of the endogenous promoter, we visualized in vivo internalization of this native GPCR upon physiological stimulation. We developed a paradigm in which animals were made dependent on morphine in a drug-paired context. When re-exposed to this context in a drug-free state, mice showed context-dependent withdrawal signs and activation of the hippocampus. Receptor internalization was transiently detected in a subset of CA1 neurons, uncovering regionally restricted opioid peptide release. Importantly, a pool of surface receptors always remained, which contrasts with the in vivo profile previously established for exogenous drug-induced internalization. Therefore, a distinct response is observed at the receptor level upon a physiological or pharmacological stimulation. Altogether, direct in vivo GPCR visualization enables mapping receptor stimulation promoted by a behavioral challenge and represents a powerful approach to study endogenous GPCR physiology.


Subject(s)
Hippocampus/metabolism , Protein Transport , Receptors, Opioid, delta/metabolism , Animals , Enkephalin, Methionine/metabolism , Female , Gene Knock-In Techniques , Hippocampus/drug effects , Male , Mice , Mice, Inbred C57BL , Molecular Imaging , Morphine/pharmacology , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/genetics , Substance Withdrawal Syndrome/metabolism
2.
Sci Immunol ; 8(90): eadf4699, 2023 Dec 22.
Article in English | MEDLINE | ID: mdl-38134241

ABSTRACT

Immune cells sense the microenvironment to fine-tune their inflammatory responses. Patients with cryopyrin-associated periodic syndrome (CAPS), caused by mutations in the NLRP3 gene, develop autoinflammation triggered by nonantigenic cues such as from the environment. However, the underlying mechanisms are poorly understood. Here, we uncover that KCNN4, a calcium-activated potassium channel, links PIEZO-mediated mechanotransduction to NLRP3 inflammasome activation. Yoda1, a PIEZO1 agonist, lowered the threshold for NLRP3 inflammasome activation. PIEZO-mediated sensing of stiffness and shear stress increased NLRP3-dependent inflammation. Myeloid-specific deletion of PIEZO1/2 protected mice from gouty arthritis. Mechanistically, activation of PIEZO1 triggers calcium influx, which activates KCNN4 to evoke potassium efflux and promotes NLRP3 inflammasome activation. Activation of PIEZO signaling was sufficient to activate the inflammasome in cells expressing CAPS-causing NLRP3 mutants via KCNN4. Last, pharmacological inhibition of KCNN4 alleviated autoinflammation in cells of patients with CAPS and in mice bearing a CAPS mutation. Thus, PIEZO-dependent mechanical inputs boost inflammation in NLRP3-dependent diseases, including CAPS.


Subject(s)
Cryopyrin-Associated Periodic Syndromes , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , Animals , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism , Mechanotransduction, Cellular , Cryopyrin-Associated Periodic Syndromes/genetics , Inflammation , Intermediate-Conductance Calcium-Activated Potassium Channels , Ion Channels/genetics
3.
Nat Metab ; 5(6): 1045-1058, 2023 06.
Article in English | MEDLINE | ID: mdl-37277610

ABSTRACT

Hypothalamic AgRP/NPY neurons are key players in the control of feeding behaviour. Ghrelin, a major orexigenic hormone, activates AgRP/NPY neurons to stimulate food intake and adiposity. However, cell-autonomous ghrelin-dependent signalling mechanisms in AgRP/NPY neurons remain poorly defined. Here we show that calcium/calmodulin-dependent protein kinase ID (CaMK1D), a genetic hot spot in type 2 diabetes, is activated upon ghrelin stimulation and acts in AgRP/NPY neurons to mediate ghrelin-dependent food intake. Global Camk1d-knockout male mice are resistant to ghrelin, gain less body weight and are protected against high-fat-diet-induced obesity. Deletion of Camk1d in AgRP/NPY, but not in POMC, neurons is sufficient to recapitulate above phenotypes. In response to ghrelin, lack of CaMK1D attenuates phosphorylation of CREB and CREB-dependent expression of the orexigenic neuropeptides AgRP/NPY in fibre projections to the paraventricular nucleus (PVN). Hence, CaMK1D links ghrelin action to transcriptional control of orexigenic neuropeptide availability in AgRP neurons.


Subject(s)
Diabetes Mellitus, Type 2 , Ghrelin , Mice , Animals , Male , Ghrelin/metabolism , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Diabetes Mellitus, Type 2/metabolism , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Neurons/metabolism , Obesity/metabolism , Mice, Knockout , Eating , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism
4.
J Neurosci ; 30(49): 16459-68, 2010 Dec 08.
Article in English | MEDLINE | ID: mdl-21147985

ABSTRACT

δ-Opioid receptors are G-protein-coupled receptors that regulate nociceptive and emotional responses. It has been well established that distinct agonists acting at the same G-protein-coupled receptor can engage different signaling or regulatory responses. This concept, known as biased agonism, has important biological and therapeutic implications. Ligand-biased responses are well described in cellular models, however, demonstrating the physiological relevance of biased agonism in vivo remains a major challenge. The aim of this study was to investigate the long-term consequences of ligand-biased trafficking of the δ-opioid receptor, at both the cellular and behavioral level. We used δ agonists with similar binding and analgesic properties, but high [SNC80 ((+)-4-[(αR)-α-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide)]- or low [ARM390 (N,N-diethyl-4-(phenyl-piperidin-4-ylidenemethyl)-benzamide)]-internalization potencies. As we found previously, a single SNC80-but not ARM390-administration triggered acute desensitization of the analgesic response in mice. However, daily injections of either compound over 5 d produced full analgesic tolerance. SNC80-tolerant animals showed widespread receptor downregulation, and tolerance to analgesic, locomotor and anxiolytic effects of the agonist. Hence, internalization-dependent tolerance developed, as a result of generalized receptor degradation. In contrast, ARM390-tolerant mice showed intact receptor expression, but δ-opioid receptor coupling to Ca²+ channels was abolished in dorsal root ganglia. Concomitantly, tolerance developed for agonist-induced analgesia, but not locomotor or anxiolytic responses. Therefore, internalization-independent tolerance was produced by anatomically restricted adaptations leading to pain-specific tolerance. Hence, ligand-directed receptor trafficking of the δ-opioid receptor engages distinct adaptive responses, and this study reveals a novel aspect of biased agonism in vivo.


Subject(s)
Analgesics/pharmacology , Drug Tolerance/physiology , Ligands , Pain Threshold/physiology , Receptors, Opioid, delta/metabolism , Analgesics/therapeutic use , Animals , Benzamides/pharmacology , Benzamides/therapeutic use , Brain/ultrastructure , Calcium/metabolism , Cell Membrane/drug effects , Cell Membrane/genetics , Disease Models, Animal , Drug Interactions , Drug Tolerance/genetics , Female , Freund's Adjuvant , Ganglia, Spinal/cytology , Green Fluorescent Proteins/genetics , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Inflammation/chemically induced , Inflammation/complications , Locomotion/drug effects , Locomotion/genetics , Male , Maze Learning/drug effects , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pain/drug therapy , Pain/etiology , Pain Threshold/drug effects , Patch-Clamp Techniques/methods , Piperazines/pharmacology , Piperazines/therapeutic use , Piperidines/pharmacology , Piperidines/therapeutic use , Protein Binding/drug effects , Protein Transport/genetics , Protein Transport/physiology , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/genetics , Sensory Receptor Cells/drug effects , Spinal Cord/ultrastructure , Statistics, Nonparametric , Sulfur Isotopes/metabolism , Time Factors
5.
Nat Commun ; 10(1): 3312, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31346174

ABSTRACT

Compromised function of insulin-secreting pancreatic ß cells is central to the development and progression of Type 2 Diabetes (T2D). However, the mechanisms underlying ß cell failure remain incompletely understood. Here, we report that metabolic stress markedly enhances macroautophagy-independent lysosomal degradation of nascent insulin granules. In different model systems of diabetes including of human origin, stress-induced nascent granule degradation (SINGD) contributes to loss of insulin along with mammalian/mechanistic Target of Rapamycin (mTOR)-dependent suppression of macroautophagy. Expression of Protein Kinase D (PKD), a negative regulator of SINGD, is reduced in diabetic ß cells. Pharmacological activation of PKD counters SINGD and delays the onset of T2D. Conversely, inhibition of PKD exacerbates SINGD, mitigates insulin secretion and accelerates diabetes. Finally, reduced levels of lysosomal tetraspanin CD63 prevent SINGD, leading to increased insulin secretion. Overall, our findings implicate aberrant SINGD in the pathogenesis of diabetes and suggest new therapeutic strategies to prevent ß cell failure.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Lysosomes/metabolism , Animals , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Humans , Insulin/chemistry , Insulin Secretion , Insulin-Secreting Cells/cytology , Macroautophagy , Male , Mice, Inbred C57BL , Protein Kinase C/genetics , Protein Kinase C/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
6.
Cell Death Dis ; 7(10): e2411, 2016 10 13.
Article in English | MEDLINE | ID: mdl-27735945

ABSTRACT

Adaptation to changes in nutrient availability is crucial for cells and organisms. Posttranslational modifications of signaling proteins are very dynamic and are therefore key to promptly respond to nutrient deprivation or overload. Herein we screened for ubiquitylation of proteins in the livers of fasted and refed mice using a comprehensive systemic proteomic approach. Among 1641 identified proteins, 117 were differentially ubiquitylated upon fasting or refeeding. Endoplasmic reticulum (ER) and secretory proteins were enriched in the livers of refed mice in part owing to an ER-stress-mediated response engaging retro-translocation and ubiquitylation of proteins from the ER. Complement C3, an innate immune factor, emerged as the most prominent ER-related hit of our screen. Accordingly, we found that secretion of C3 from the liver and primary hepatocytes as well as its dynamic trafficking are nutrient dependent. Finally, obese mice with a chronic nutrient overload show constitutive trafficking of C3 in the livers despite acute changes in nutrition, which goes in line with increased C3 levels and low-grade inflammation reported for obese patients. Our study thus suggests that nutrient sensing in the liver is coupled to release of C3 and potentially its metabolic and inflammatory functions.


Subject(s)
Complement C3/metabolism , Liver/metabolism , Proteome/metabolism , Ubiquitins/metabolism , Animals , Endoplasmic Reticulum/metabolism , Feeding Behavior , HEK293 Cells , Humans , Intracellular Space/metabolism , Mice, Inbred C57BL , Protein Transport , Secretory Vesicles/metabolism , Stress, Physiological , Ubiquitination
7.
J Neurosci ; 24(38): 8214-22, 2004 Sep 22.
Article in English | MEDLINE | ID: mdl-15385604

ABSTRACT

Reduced glutamate-mediated synaptic transmission has been implicated in the pathophysiology of schizophrenia. Because antipsychotic agents might exert their beneficial effects against schizophrenic symptoms by strengthening excitatory transmission in critical dopaminoceptive brain areas, in the present study we have studied the effects of acute and chronic haloperidol treatment on striatal synaptic plasticity. Repetitive stimulation of corticostriatal terminals in slices induced either long-term depression or long-term potentiation (LTP) of excitatory transmission in control rats, whereas it invariably induced NMDA receptor-dependent LTP in animals treated chronically with haloperidol. Haloperidol effects were mimicked and occluded in mice lacking both D2L and D2S isoforms of dopamine D2 receptors (D2R-/-), in mice lacking D2L receptors and expressing normal levels of D2S receptors (D2R-/-;D2L-/-), and in mice lacking D2L receptors and overexpressing D2S receptors (D2L-/-). These data indicate that the blockade of D2L receptors was responsible for the LTP-favoring action of haloperidol in the striatum. In contrast, overexpression of D2S receptors uncovered a facilitatory role of this receptor isoform in LTP formation because LTP recorded from D2L-/- mice, but not those recorded from wild-type, D2R-/-, and D2R-/-;D2L-/- mice, was insensitive to the pharmacological blockade of D1 receptors. The identification of the cellular, molecular, and receptor mechanisms involved in the action of haloperidol in the brain is essential to understand how antipsychotic agents exert their beneficial and side effects.


Subject(s)
Cerebral Cortex/drug effects , Haloperidol/pharmacology , Long-Term Potentiation/drug effects , Neostriatum/drug effects , Receptors, Dopamine D2/drug effects , Animals , Antipsychotic Agents/pharmacology , Binding Sites/drug effects , Cerebral Cortex/physiology , Dopamine Antagonists/pharmacology , Ligands , Long-Term Potentiation/physiology , Long-Term Synaptic Depression/drug effects , Long-Term Synaptic Depression/physiology , Male , Mice , Mice, Knockout , Motor Activity/drug effects , Neostriatum/physiology , Neuronal Plasticity/drug effects , Organ Culture Techniques , Patch-Clamp Techniques , Protein Kinase C/drug effects , Protein Kinase C/physiology , Rats , Rats, Wistar , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/physiology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Time Factors
8.
J Neurosci ; 23(15): 6245-54, 2003 Jul 16.
Article in English | MEDLINE | ID: mdl-12867509

ABSTRACT

By stimulating distinct receptor subtypes, dopamine (DA) exerts presynaptic and postsynaptic actions on both large aspiny (LA) cholinergic and fast-spiking (FS) parvalbumin-positive interneurons of the striatum. Lack of receptor- and isoform-specific pharmacological agents, however, has hampered the progress toward a detailed identification of the specific DA receptors involved in these actions. To overcome this issue, in the present study we used four different mutant mice in which the expression of specific DA receptors was ablated. In D1 receptor null mice, D1R-/-, DA dose-dependently depolarized both LA and FS interneurons. Interestingly, SCH 233390 (10 microm), a D1-like (D1 and D5) receptor antagonist, but not l-sulpiride (3-10 microm), a D2-like (D2, D3, D4) receptor blocker, prevented this effect, implying D5 receptors in this action. Accordingly, immunohistochemical analyses in both wild-type and D1R-/- mice confirmed the expression of D5 receptors in both cholinergic and parvalbumin-positive interneurons of the striatum. In mice lacking D2 receptors, D2R-/-, the DA-dependent inhibition of GABA transmission was lost in both interneuron populations. Both isoforms of D2 receptor, D2L and D2S, were very likely involved in this inhibitory action, as revealed by the electrophysiological analysis of the effect of the DA D2-like receptor agonist quinpirole in two distinct mutants lacking D2L receptors and expressing variable contents of D2S receptors. The identification of the receptor subtypes involved in the actions of DA on different populations of striatal cells is essential to understand the circuitry of the basal ganglia and to develop pharmacological strategies able to interfere selectively with specific neuronal functions.


Subject(s)
Corpus Striatum/metabolism , Dopamine/pharmacology , Interneurons/metabolism , Receptors, Dopamine/metabolism , Synapses/metabolism , Animals , Antibody Specificity , Blotting, Western , Corpus Striatum/cytology , Corpus Striatum/drug effects , Dopamine Agonists/pharmacology , Electrophysiology , In Vitro Techniques , Interneurons/drug effects , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Membrane Potentials/physiology , Mice , Mice, Knockout , Patch-Clamp Techniques , Presynaptic Terminals/drug effects , Presynaptic Terminals/physiology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Dopamine/deficiency , Receptors, Dopamine/genetics , Receptors, Dopamine D1/biosynthesis , Receptors, Dopamine D1/deficiency , Receptors, Dopamine D1/genetics , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/deficiency , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D5 , Receptors, GABA-A/metabolism , Synapses/drug effects
9.
Brain Res ; 1052(1): 82-7, 2005 Aug 02.
Article in English | MEDLINE | ID: mdl-15996639

ABSTRACT

We studied the role of the dopamine D2 receptor in physiological regulation of pain-related behavior. The experiments were performed in dopamine D2 receptor knockout mice and in their wild-type controls. Baseline sensitivity to thermal nociception was determined by measuring the response latency in the hot plate at three different stimulus temperatures and by determining the radiant-heat-induced paw withdrawal. Mechanical sensitivity was assessed by determining paw withdrawal responses to stimulation with a calibrated series of monofilaments. Intracolonic capsaicin was used to produce sustained pain-related behavior and referred hypersensitivity to mechanical stimulation. The hot plate response latencies were not significantly different between the dopamine D2 receptor knockout and wild-type animals, although the stimulus temperature-dependent decrease in the response latency was steeper in the wild-type group. The radiant-heat-induced paw withdrawal latency was slightly longer in the knockout animals. The number of capsaicin-induced behavioral responses or the latency to the occurrence of the first capsaicin-induced response was not different between the experimental groups. Dopamine D2 receptor knockout animals were more sensitive to mechanical stimulation of the hindpaws than wild-type animals both in the baseline condition and following development of capsaicin-induced referred hypersensitivity in the hindpaws. The results indicate that dopamine D2 receptors influence baseline nociception in the mouse, although this effect is weak and submodality selective. Additionally, dopamine D2 receptors may contribute to attenuation of referred hypersensitivity caused by sustained nociception.


Subject(s)
Pain/physiopathology , Receptors, Dopamine D2/deficiency , Animals , Behavior, Animal , Capsaicin/toxicity , Hyperalgesia/genetics , Hyperalgesia/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pain/chemically induced , Pain/genetics , Pain Measurement , Physical Stimulation/adverse effects , Reaction Time/physiology , Receptors, Dopamine D2/physiology
10.
Science ; 347(6224): 878-82, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25700520

ABSTRACT

Pancreatic ß cells lower insulin release in response to nutrient depletion. The question of whether starved ß cells induce macroautophagy, a predominant mechanism maintaining energy homeostasis, remains poorly explored. We found that, in contrast to many mammalian cells, macroautophagy in pancreatic ß cells was suppressed upon starvation. Instead, starved ß cells induced lysosomal degradation of nascent secretory insulin granules, which was controlled by protein kinase D (PKD), a key player in secretory granule biogenesis. Starvation-induced nascent granule degradation triggered lysosomal recruitment and activation of mechanistic target of rapamycin that suppressed macroautophagy. Switching from macroautophagy to insulin granule degradation was important to keep insulin secretion low upon fasting. Thus, ß cells use a PKD-dependent mechanism to adapt to nutrient availability and couple autophagy flux to secretory function.


Subject(s)
Autophagy , Insulin-Secreting Cells/physiology , Insulin/metabolism , Secretory Vesicles/physiology , Animals , Cells, Cultured , Fasting , Humans , Insulin Secretion , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/ultrastructure , Mice , Mice, Mutant Strains , Mice, Transgenic , Mitogen-Activated Protein Kinase 13/genetics , Protein Kinase C/physiology , Secretory Vesicles/metabolism
11.
Brain Struct Funct ; 220(2): 677-702, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24623156

ABSTRACT

Opioid receptors are G protein-coupled receptors (GPCRs) that modulate brain function at all levels of neural integration, including autonomic, sensory, emotional and cognitive processing. Mu (MOR) and delta (DOR) opioid receptors functionally interact in vivo, but whether interactions occur at circuitry, cellular or molecular levels remains unsolved. To challenge the hypothesis of MOR/DOR heteromerization in the brain, we generated redMOR/greenDOR double knock-in mice and report dual receptor mapping throughout the nervous system. Data are organized as an interactive database offering an opioid receptor atlas with concomitant MOR/DOR visualization at subcellular resolution, accessible online. We also provide co-immunoprecipitation-based evidence for receptor heteromerization in these mice. In the forebrain, MOR and DOR are mainly detected in separate neurons, suggesting system-level interactions in high-order processing. In contrast, neuronal co-localization is detected in subcortical networks essential for survival involved in eating and sexual behaviors or perception and response to aversive stimuli. In addition, potential MOR/DOR intracellular interactions within the nociceptive pathway offer novel therapeutic perspectives.


Subject(s)
Brain/metabolism , Nerve Net/metabolism , Neurons/metabolism , Receptors, Opioid, delta/analysis , Receptors, Opioid, mu/analysis , Animals , Female , Gene Knock-In Techniques , Male , Mice , Mice, Inbred C57BL
12.
Article in English | MEDLINE | ID: mdl-25938125

ABSTRACT

Opioid receptors belong to the G protein coupled receptor family. They modulate brain function at all levels of neural integration and therefore impact on autonomous, sensory, emotional and cognitive processing. In vivo functional interaction between mu and delta opioid receptors are known to take place though it is still debated whether interactions occur at circuitry, cellular or molecular level. Also, the notion of receptor crosstalk via mu-delta heteromers is well documented in vitro but in vivo evidence remains scarce. To identify neurons in which receptor interactions could take place, we designed a unique double mutant knock-in mouse line that expresses functional red-fluorescent mu receptors and green-fluorescent delta receptors. We mapped mu and delta receptor distribution and co-localization throughout the nervous system and created the first interactive brain atlas with concomitant mu-delta visualization at subcellular resolution (http://mordor.ics-mci.fr/). Mu and delta receptors co-localize in neurons from subcortical networks but are mainly detected in separate neurons in the forebrain. Also, co-immunoprecipitation experiments indicated physical proximity in the hippocampus, a prerequisite to mu-delta heteromerization. Altogether, data suggest that mu-delta functional interactions take place at systems level for high-order emotional and cognitive processing whereas mu-delta may interact at cellular level in brain networks essential for survival, which has potential implications for innovative drug design in pain control, drug addiction and eating disorders.

13.
J Biotechnol ; 166(4): 182-6, 2013 Jul 20.
Article in English | MEDLINE | ID: mdl-23732834

ABSTRACT

In vivo conditional knock-out of a protein is a method of choice to decipher its biological function. It can be achieved by encoding the cre-recombinase on a recombinant virus to exert spatio-temporal control of its expression and enzymatic activity and, subsequently, of the target gene deletion. Recombinant baculoviruses have been successfully used to express a wide range of proteins in insect cells. More recently, their potential to infect mammalian cells has been addressed but, so far, their ability to yield a conditional knock-out as a result of efficient in vivo cre-recombinase gene delivery has not been examined. Cre-recombinase fused to the green fluorescent protein was cloned under the control of the CAG promoter in a recombinant Autographa californica baculovirus expressing the vesicular stomatitis virus envelope G protein for increased mammalian cell infection. Gene delivery was evaluated in vitro in mammalian cells, neuroblastoma and mouse primary neuronal cultures as well as in vivo in the mouse brain. Infection with adeno-associated viruses encoding the cre-recombinase fused to the green fluorescent protein was performed as a positive control. Our results indicate that baculovirus infection leads to functional cre-recombinase expression in non-neuronal and neuroblastoma cell lines but not in mouse primary neuronal cultures or brain.


Subject(s)
Baculoviridae , Gene Transfer Techniques , Integrases/genetics , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Dependovirus/genetics , Dependovirus/metabolism , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , HEK293 Cells , Humans , Mice , Parvoviridae Infections/genetics
14.
Dev Cell ; 23(4): 756-68, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-22981988

ABSTRACT

BAR domains can prevent membrane fission through their ability to shield necks of budding vesicles from fission-inducing factors. However, the physiological role of this inhibitory function and its regulation is unknown. Here we identify a checkpoint involving the BAR-domain-containing protein Arfaptin-1 that controls biogenesis of secretory granules at the trans-Golgi network (TGN). We demonstrate that protein kinase D (PKD) phosphorylates Arfaptin-1 at serine 132, which disrupts the ability of Arfaptin-1 to inhibit the activity of ADP ribosylation factor, an important component of the vesicle scission machinery. The physiological significance of this regulatory mechanism is evidenced by loss of glucose-stimulated insulin secretion due to granule scission defects in pancreatic ß cells expressing nonphosphorylatable Arfaptin-1. Accordingly, depletion of Arfaptin-1 leads to the generation of small nonfunctional secretory granules. Hence, PKD-mediated Arfaptin-1 phosphorylation is necessary to ensure biogenesis of functional transport carriers at the TGN in regulated secretion.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Secretory Vesicles/metabolism , trans-Golgi Network/metabolism , ADP-Ribosylation Factors/antagonists & inhibitors , ADP-Ribosylation Factors/metabolism , Animals , Cell Line, Tumor , Phosphorylation , Protein Kinase C/metabolism , Protein Structure, Tertiary , Rats , Serine/metabolism
15.
Proc Natl Acad Sci U S A ; 101(1): 325-30, 2004 Jan 06.
Article in English | MEDLINE | ID: mdl-14694200

ABSTRACT

The role played by oligodendrocytes (OLs), the myelinating cells of the CNS, during brain development has not been fully explored. We have addressed this question by inducing a temporal and reversible ablation of OLs on postnatal CNS development. OL ablation in newborn mice leads to a profound alteration in the structure of the cerebellar cortex, which can be progressively rescued by newly generated cells, leading to a delayed myelination. Nevertheless, the temporal shift of the OL proliferation and myelinating program cannot completely compensate for developmental defects, resulting in impaired motor functions in the adult. Strikingly, we show that, despite these abnormalities, epigenetic factors, such as motor training, are able to fully rescue cerebellar-directed motor skills.


Subject(s)
Cerebellar Cortex/physiopathology , Motor Skills/physiology , Oligodendroglia/physiology , Physical Conditioning, Animal/physiology , Animals , Animals, Newborn , Cerebellar Cortex/growth & development , Cerebellar Cortex/ultrastructure , Demyelinating Diseases/physiopathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron , Myelin Basic Protein/genetics , Myelin Sheath/physiology , R-SNARE Proteins , Synaptic Vesicles/ultrastructure , Thymidine Kinase/genetics
16.
Proc Natl Acad Sci U S A ; 100(7): 4305-9, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12651945

ABSTRACT

Dopamine D2 receptors are highly expressed in the dorsal striatum where they participate in the regulation of (i) tyrosine hydroxylase (TH), in nigrostriatal nerve terminals, and (ii) the dopamine- and cAMP-regulated phosphoprotein of 32 kDa (DARPP-32), in medium spiny neurons. Two isoforms of the D2 receptor are generated by differential splicing of the same gene and are referred to as short (D2S) and long (D2L) dopamine receptors. Here we have used wild-type mice, dopamine D2 receptor knockout mice (D2 KO mice; lacking both D2S and D2L receptors) and D2L receptor-selective knockout mice (D2L KO mice) to evaluate the involvement of each isoform in the regulation of the phosphorylation of TH and DARPP-32. Incubation of striatal slices from wild-type mice with quinpirole, a dopamine D2 receptor agonist, decreased the state of phosphorylation of TH at Ser-40 and its enzymatic activity. Both effects were abolished in D2 KO mice but were still present in D2L KO mice. In wild-type mice, quinpirole inhibits the increase in DARPP-32 phosphorylation at Thr-34 induced by SKF81297, a dopamine D1 receptor agonist. This effect is absent in D2 KO as well as D2L KO mice. The inability of quinpirole to regulate DARPP-32 phosphorylation in D2L KO mice cannot be attributed to decreased coupling of D2S receptors to G proteins, because quinpirole produces a similar stimulation of [(35)S]GTPgammaS binding in wild-type and D2L KO mice. These results demonstrate that D2S and D2L receptors participate in presynaptic and postsynaptic dopaminergic transmission, respectively.


Subject(s)
Corpus Striatum/physiology , Dopamine Agonists/pharmacology , Nerve Tissue Proteins/metabolism , Phosphoproteins/metabolism , Receptors, Dopamine D2/physiology , Synapses/physiology , Animals , Corpus Striatum/drug effects , Dopamine and cAMP-Regulated Phosphoprotein 32 , Female , Homeostasis , Mice , Mice, Knockout , Phosphorylation , Protein Isoforms/physiology , Quinpirole/pharmacology , Receptors, Dopamine D2/deficiency , Receptors, Dopamine D2/genetics , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL