229 research outputs found

    Colicin N and its thermolytic fragment induce phospholipid vesicle fusion

    Get PDF
    AbstractColicin N, a bacteriocin encoded on a plasmid belonging to the pore-forming class of colicins, induces phospholipid vesicle fusion at acidic pH as demonstrated by fluorescence resonance energy transfer. Its C-terminal thermolytic fragment has properties very similar to the native molecule. The fusion is protein concentration-dependent and is regulated by (a) group(s) with a pK of approximately 4.6. The physiological relevance of this characteristic common to all colicins tested so far is discussed

    G protein-coupled receptor heteromers are key players in substance use disorder

    Get PDF
    International audienceG protein-coupled receptors (GPCR) represent the largest family of membrane proteins in the human genome. Physical association between two different GPCRs is linked to functional interactions which generates a novel entity, called heteromer, with specific ligand binding and signaling properties. Heteromerization is increasingly recognized to take place in the mesocorticolimbic pathway and to contribute to various aspects related to substance use disorder. This review focuses on heteromers identified in brain areas relevant to drug addiction. We report changes at the molecular and cellular levels that establish specific functional impact and highlight behavioral outcome in preclinical models. Finally, we briefly discuss selective targeting of native heteromers as an innovative therapeutic option

    Enhanced spontaneous activity of the mu opioid receptor by cysteine mutations: characterization of a tool for inverse agonist screening.

    Get PDF
    BACKGROUND: The concept of spontaneous- or constitutive-activity has become widely accepted and verified for numerous G protein-coupled receptors and this ligand-independent activity is also acknowledged to play a role in some pathologies. Constitutive activity has been reported for the mu opioid receptor. In some cases the increase in receptor basal activity was induced by chronic morphine administration suggesting that constitutive activity may contribute to the development of drug tolerance and dependence. Constitutively active mutants represent excellent tools for gathering information about the mechanisms of receptor activation and the possible physiological relevance of spontaneous receptor activity. The high basal level of activity of these mutants also allows for easier identification of inverse agonists, defined as ligands able to suppress spontaneous receptor activity, and leads to a better comprehension of their modulatory effects as well as possible in vivo use. RESULTS: Cysteines 348 and 353 of the human mu opioid receptor (hMOR) were mutated into alanines and Ala(348,353 )hMOR was stably expressed in HEK 293 cells. [(35)S] GTPγS binding experiments revealed that Ala(348,353 )hMOR basal activity was significantly higher when compared to hMOR, suggesting that the mutant receptor is constitutively active. [(35)S] GTPγS binding was decreased by cyprodime or CTOP indicating that both ligands have inverse agonist properties. All tested agonists exhibited binding affinities higher for Ala(348,353 )hMOR than for hMOR, with the exception of endogenous opioid peptides. Antagonist affinity remained virtually unchanged except for CTOP and cyprodime that bound the double mutant with higher affinities. The agonists DAMGO and morphine showed enhanced potency for the Ala(348,353 )hMOR receptor in [(35)S] GTPγS experiments. Finally, pretreatment with the antagonists naloxone, cyprodime or CTOP significantly increased Ala(348,353 )hMOR expression. CONCLUSION: Taken together our data indicate that the double C348/353A mutation results in a constitutively active conformation of hMOR that is still activated by agonists. This is the first report of a stable CAM of hMOR with the potential to screen for inverse agonists

    Heteromerization Modulates mu Opioid Receptor Functional Properties in vivo.

    Get PDF
    Mu opioid receptors modulate a large number of physiological functions. They are in particular involved in the control of pain perception and reward properties. They are also the primary molecular target of opioid drugs and mediate their beneficial analgesic effects, euphoric properties as well as negative side effects such as tolerance and physical dependence. Importantly, mu opioid receptors can physically associate with another receptor to form a novel entity called heteromer that exhibits specific ligand binding, signaling, and trafficking properties. As reviewed here, in vivo physical proximity has now been evidenced for several receptor pairs, subsequent impact of heteromerization on native mu opioid receptor signaling and trafficking identified and a link to behavioral changes established. Selective targeting of heteromers as a tool to modulate mu opioid receptor activity is therefore attracting growing interest and raises hopes for innovative therapeutic strategies.journal articlereview20182018 11 13importe

    Double fluorescent knock-in mice to investigate endogenous mu-delta opioid heteromer subscellular distribution.

    Get PDF
    The heteromerization of Mu (MOP) and delta (DOP) opioid receptors has been extensively studied in heterologous systems. These studies demonstrated significant functional interaction of MOP and DOP evidenced by new pharmacological properties and intracellular signalling in transfected cells co-expressing the receptors. Due to the lack of appropriate tools for receptor visualization, such as specific antibodies, the pharmacological and functional properties of MOP-DOP heteromers in cells naturally expressing these receptors remains poorly understood. To address endogenous MOP-DOP heteromer trafficking and signalling in vivo and in primary neuronal cultures, we generated a double knock-in mouse line expressing functional fluorescent versions of DOP and MOP receptors. This mouse model has successfully been used to map the neuroanatomic distribution of the receptors and to identify brain regions in which the MOP-DOP heteromers are expressed. Here, we describe a method to quantitatively and automatically analyze changes in the subcellular distribution of MOP-DOP heteromers in primary hippocampal culture from this mouse model. This approach provides a unique tool to address specificities of endogenous MOP-DOP heteromer trafficking

    In vivo neuronal co-expression of mu and delta opioid receptors uncovers new therapeutic perspectives

    Get PDF
    Opioid receptors are G protein coupled receptors that modulate brain function at all levels of neural integration, including autonomous, sensory, emotional and cognitive processing. Mu and delta opioid receptors functionally interact in vivo, but whether interactions occur at circuitry, cellular or molecular level remains unsolved. 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

    How modeling can reconcile apparently discrepant experimental results: the case of pacemaking in dopaminergic neurons.

    Get PDF
    Midbrain dopaminergic neurons are endowed with endogenous slow pacemaking properties. In recent years, many different groups have studied the basis for this phenomenon, often with conflicting conclusions. In particular, the role of a slowly-inactivating L-type calcium channel in the depolarizing phase between spikes is controversial, and the analysis of slow oscillatory potential (SOP) recordings during the blockade of sodium channels has led to conflicting conclusions. Based on a minimal model of a dopaminergic neuron, our analysis suggests that the same experimental protocol may lead to drastically different observations in almost identical neurons. For example, complete L-type calcium channel blockade eliminates spontaneous firing or has almost no effect in two neurons differing by less than 1% in their maximal sodium conductance. The same prediction can be reproduced in a state of the art detailed model of a dopaminergic neuron. Some of these predictions are confirmed experimentally using single-cell recordings in brain slices. Our minimal model exhibits SOPs when sodium channels are blocked, these SOPs being uncorrelated with the spiking activity, as has been shown experimentally. We also show that block of a specific conductance (in this case, the SK conductance) can have a different effect on these two oscillatory behaviors (pacemaking and SOPs), despite the fact that they have the same initiating mechanism. These results highlight the fact that computational approaches, besides their well known confirmatory and predictive interests in neurophysiology, may also be useful to resolve apparent discrepancies between experimental results

    Heteromerization Modulates mu Opioid Receptor Functional Properties in vivo

    Get PDF
    Mu opioid receptors modulate a large number of physiological functions. They are in particular involved in the control of pain perception and reward properties. They are also the primary molecular target of opioid drugs and mediate their beneficial analgesic effects, euphoric properties as well as negative side effects such as tolerance and physical dependence. Importantly, mu opioid receptors can physically associate with another receptor to form a novel entity called heteromer that exhibits specific ligand binding, signaling, and trafficking properties. As reviewed here, in vivo physical proximity has now been evidenced for several receptor pairs, subsequent impact of heteromerization on native mu opioid receptor signaling and trafficking identified and a link to behavioral changes established. Selective targeting of heteromers as a tool to modulate mu opioid receptor activity is therefore attracting growing interest and raises hopes for innovative therapeutic strategies

    Fluorescent knock-in mice to decipher the physiopathological role of G protein-coupled receptors.

    Get PDF
    G protein-coupled receptors (GPCRs) modulate most physiological functions but are also critically involved in numerous pathological states. Approximately a third of marketed drugs target GPCRs, which places this family of receptors in the main arena of pharmacological pre-clinical and clinical research. The complexity of GPCR function demands comprehensive appraisal in native environment to collect in-depth knowledge of receptor physiopathological roles and assess the potential of therapeutic molecules. Identifying neurons expressing endogenous GPCRs is therefore essential to locate them within functional circuits whereas GPCR visualization with subcellular resolution is required to get insight into agonist-induced trafficking. Both remain frequently poorly investigated because direct visualization of endogenous receptors is often hampered by the lack of appropriate tools. Also, monitoring intracellular trafficking requires real-time visualization to gather in-depth knowledge. In this context, knock-in mice expressing a fluorescent protein or a fluorescent version of a GPCR under the control of the endogenous promoter not only help to decipher neuroanatomical circuits but also enable real-time monitoring with subcellular resolution thus providing invaluable information on their trafficking in response to a physiological or a pharmacological challenge. This review will present the animal models and discuss their contribution to the understanding of the physiopathological role of GPCRs. We will also address the drawbacks associated with this methodological approach and browse future directions.journal articlereview20142015 01 06importe

    Associations entre récepteurs opioïdes : vers de nouvelles stratégies thérapeutiques pour la douleur et l’addiction

    Get PDF
    Opioid receptors modulate numerous physiological functions. These receptors can associate each other to form a new functional entity named heteromer, which possesses specific functional properties. In vivo data suggest a crucial role for opioid heteromers in acute or chronic pain as well as addiction, pointing them as a new therapeutic target for the treatment of these pathologies
    • …
    corecore