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










Publication year range
1.
Acta Neuropathol ; 140(2): 143-167, 2020 08.
Article in English | MEDLINE | ID: mdl-32572598

ABSTRACT

Axonal loss is the key pathological substrate of neurological disability in demyelinating disorders, including multiple sclerosis (MS). However, the consequences of demyelination on neuronal and axonal biology are poorly understood. The abundance of mitochondria in demyelinated axons in MS raises the possibility that increased mitochondrial content serves as a compensatory response to demyelination. Here, we show that upon demyelination mitochondria move from the neuronal cell body to the demyelinated axon, increasing axonal mitochondrial content, which we term the axonal response of mitochondria to demyelination (ARMD). However, following demyelination axons degenerate before the homeostatic ARMD reaches its peak. Enhancement of ARMD, by targeting mitochondrial biogenesis and mitochondrial transport from the cell body to axon, protects acutely demyelinated axons from degeneration. To determine the relevance of ARMD to disease state, we examined MS autopsy tissue and found a positive correlation between mitochondrial content in demyelinated dorsal column axons and cytochrome c oxidase (complex IV) deficiency in dorsal root ganglia (DRG) neuronal cell bodies. We experimentally demyelinated DRG neuron-specific complex IV deficient mice, as established disease models do not recapitulate complex IV deficiency in neurons, and found that these mice are able to demonstrate ARMD, despite the mitochondrial perturbation. Enhancement of mitochondrial dynamics in complex IV deficient neurons protects the axon upon demyelination. Consequently, increased mobilisation of mitochondria from the neuronal cell body to the axon is a novel neuroprotective strategy for the vulnerable, acutely demyelinated axon. We propose that promoting ARMD is likely to be a crucial preceding step for implementing potential regenerative strategies for demyelinating disorders.


Subject(s)
Demyelinating Diseases/pathology , Mitochondria/pathology , Multiple Sclerosis/pathology , Nerve Degeneration/pathology , Neuroprotection/physiology , Animals , Axons/pathology , Humans , Mice , Organelle Biogenesis
2.
Neurobiol Dis ; 142: 104961, 2020 08.
Article in English | MEDLINE | ID: mdl-32531343

ABSTRACT

Effective analgesic treatment for neuropathic pain remains an unmet need, so previous evidence that epidermal growth factor receptor inhibitors (EGFRIs) provide unexpected rapid pain relief in a clinical setting points to a novel therapeutic opportunity. The present study utilises rodent models to address the cellular and molecular basis for the findings, focusing on primary sensory neurons because clinical pain relief is provided not only by small molecule EGFRIs, but also by the anti-EGFR antibodies cetuximab and panitumumab, which are unlikely to access the central nervous system in therapeutic concentrations. We report robust, rapid and dose-dependent analgesic effects of EGFRIs in two neuropathic pain models, matched by evidence with highly selective antibodies that expression of the EGFR (ErbB1 protein) is limited to small nociceptive afferent neurons. As other ErbB family members can heterodimerise with ErbB1, we investigated their distribution, showing consistent co-expression of ErbB2 but not ErbB3 or ErbB4, with ErbB1 in cell bodies of nociceptors, as well as providing evidence for direct molecular interaction of ErbB1 with ErbB2 in situ. Co-administration of selective ErbB1 and ErbB2 inhibitors produced clear evidence of greater-than-additive, synergistic analgesia; highlighting the prospect of a unique new combination therapy in which enhanced efficacy could be accompanied by minimisation of side-effects. Peripheral (intraplantar) administration of EGF elicited hypersensitivity only following nerve injury and this was reversed by local co-administration of selective inhibitors of either ErbB1 or ErbB2. Investigating how ErbB1 is activated in neuropathic pain, we found evidence for a role of Src tyrosine kinase, which can be activated by signals from inflammatory mediators, chemokines and cytokines during neuroinflammation. Considering downstream consequences of ErbB1 activation in neuropathic pain, we found direct recruitment to ErbB1 of an adapter for PI 3-kinase and Akt signalling together with clear Akt activation and robust analgesia from selective Akt inhibitors. The known Akt target and regulator of vesicular trafficking, AS160 was strongly phosphorylated at a perinuclear location during neuropathic pain in an ErbB1-, ErbB2- and Akt-dependent manner, corresponding to clustering and translocation of an AS160-partner, the vesicular chaperone, LRP1. Exploring whether neuronal ion channels that could contribute to hyperexcitability might be transported by this vesicular trafficking pathway we were able to identify Nav1.9, (Nav1.8) and Cav1.2 moving towards the plasma membrane or into proximal axonal locations - a process prevented by ErbB1 or Akt inhibitors. Overall these findings newly reveal both upstream and downstream signals to explain how ErbB1 can act as a signalling hub in neuropathic pain models and identify the trafficking of key ion channels to neuronal subcellular locations likely to contribute to hyperexcitability. The new concept of combined treatment with ErbB1 plus ErbB2 blockers is mechanistically validated as a promising strategy for the relief of neuropathic pain.


Subject(s)
ErbB Receptors/metabolism , Neuralgia/metabolism , Nociceptors/metabolism , Animals , Mice , Neuralgia/chemically induced , Oxaliplatin , Phosphorylation , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
3.
Pain Pract ; 20(8): 859-877, 2020 11.
Article in English | MEDLINE | ID: mdl-32474998

ABSTRACT

OBJECTIVES: Responses of spinal progenitors to spinal cord stimulation (SCS) following spinal cord injury (SCI) in rats were assessed to reveal their potential contribution to SCS-induced analgesia. METHODS: Spinal epidural electrodes were implanted in rats at T12 rostral to a quadrant dorsal horn injury at T13. Further groups additionally received either a microlesion to the dorsolateral funiculus (DLF) or gabapentin (10 mg/kg). SCS was performed at 25 Hz for 10 minutes on day 4 (early SCS) and at 10 Hz for 10 minutes on day 8 (late SCS) after injury. Paw withdrawal threshold (PWT) was measured before injury, 30 minutes before or after SCS, and before cull on day 14, followed by immunostaining assessment. RESULTS: Paw withdrawal thresholds in uninjured animals (51.0 ± 4.0 g) were markedly reduced after SCI (17.3 ± 2.2 g). This was significantly increased by early SCS (38.5 ± 5.2 g, P < 0.01) and further enhanced by late SCS (50.9 ± 1.9 g, P < 0.01) over 6 days. Numbers of neural progenitors expressing nestin, Sox2, and doublecortin (DCX) in the spinal dorsal horn were increased 6 days after SCS by 6-fold, 2-fold, and 2.5-fold, respectively (P < 0.05 to 0.01). The elevated PWT evoked by SCS was abolished by DLF microlesions (48.9 ± 2.6 g vs. 19.0 ± 3.9 g, P < 0.01) and the number of nestin-positive cells was reduced to the level without SCS (P < 0.05). Gabapentin enhanced late SCS-induced analgesia from 37.0 ± 3.9 g to 54.0 ± 0.8 g (P < 0.01) and increased gamma-aminobutyric acid (GABA)-ergic neuronal marker vesicular GABA transporter-positive newborn cells 2-fold (P < 0.01). CONCLUSIONS: Spinal progenitor cells appear to be activated by SCS via descending pathways, which may be enhanced by gabapentin and potentially contributes to relief of SCI-induced neuropathic pain.


Subject(s)
Neural Stem Cells/physiology , Neuralgia/physiopathology , Spinal Cord Injuries/physiopathology , Spinal Cord Stimulation , Analgesia/methods , Animals , Doublecortin Protein , Hypesthesia/etiology , Hypesthesia/physiopathology , Male , Neuralgia/etiology , Pain Management/methods , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/complications
4.
Pain Med ; 20(12): 2495-2505, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31106835

ABSTRACT

OBJECTIVE: Case reports and a case series have described relief of neuropathic pain (NP) after treatment with epidermal growth factor receptor inhibitors (EGFR-Is). These observations are supported by preclinical findings. The aim of this trial was to explore a potential clinical signal supporting the therapeutic efficacy of EGFR-Is in NP. METHODS: In a proof-of-concept trial using a randomized, double-blind, placebo-controlled design, 14 patients with severe, chronic, therapy-resistant NP due to compressed peripheral nerves or complex regional pain syndrome were randomized to receive a single infusion of the EGFR-I cetuximab and placebo in crossover design, followed by a single open-label cetuximab infusion. RESULTS: The mean reduction in daily average pain scores three to seven days after single-blinded cetuximab infusion was 1.73 points (90% confidence interval [CI] = 0.80 to 2.66), conferring a 1.22-point greater reduction than placebo (90% CI = -0.10 to 2.54). Exploratory analyses suggested that pain reduction might be greater in the 14 days after treatment with blinded cetuximab than after placebo. The proportion of patients who reported ≥50% reduction in average pain three to seven days after cetuximab was 36% (14% after placebo), and comparison of overall pain reduction suggests a trend in favor of cetuximab. Skin rash (grade 1-2) was the most frequent side effect (12/14, 86%). CONCLUSIONS: This small proof-of-concept evaluation of an EGFR-I against NP did not provide statistical evidence of efficacy. However, substantial reductions in pain were reported, and confidence intervals do not rule out a clinically meaningful treatment effect. Evaluation of EGFR-I against NP therefore warrants further investigation.


Subject(s)
Cetuximab/therapeutic use , Complex Regional Pain Syndromes/drug therapy , ErbB Receptors/antagonists & inhibitors , Nerve Compression Syndromes/drug therapy , Neuralgia/drug therapy , Adult , Double-Blind Method , Female , Humans , Male , Middle Aged , Pain Measurement , Proof of Concept Study , Treatment Outcome , Young Adult
5.
Mol Neurobiol ; 56(8): 5917-5933, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30689196

ABSTRACT

As mitochondrial dysfunction is evident in neurodegenerative disorders that are accompanied by pain, we generated inducible mutant mice with disruption of mitochondrial respiratory chain complex IV, by COX10 deletion limited to sensory afferent neurons through the use of an Advillin Cre-reporter. COX10 deletion results in a selective energy-deficiency phenotype with minimal production of reactive oxygen species. Mutant mice showed reduced activity of mitochondrial respiratory chain complex IV in many sensory neurons, increased ADP/ATP ratios in dorsal root ganglia and dorsal spinal cord synaptoneurosomes, as well as impaired mitochondrial membrane potential, in these synaptoneurosome preparations. These changes were accompanied by marked pain hypersensitivity in mechanical and thermal (hot and cold) tests without altered motor function. To address the underlying basis, we measured Ca2+ fluorescence responses of dorsal spinal cord synaptoneurosomes to activation of the GluK1 (kainate) receptor, which we showed to be widely expressed in small but not large nociceptive afferents, and is minimally expressed elsewhere in the spinal cord. Synaptoneurosomes from mutant mice showed greatly increased responses to GluK1 agonist. To explore whether altered nucleotide levels may play a part in this hypersensitivity, we pharmacologically interrogated potential roles of AMP-kinase and ADP-sensitive purinergic receptors. The ADP-sensitive P2Y1 receptor was clearly implicated. Its expression in small nociceptive afferents was increased in mutants, whose in vivo pain hypersensitivity, in mechanical, thermal and cold tests, was reversed by a selective P2Y1 antagonist. Energy depletion and ADP elevation in sensory afferents, due to mitochondrial respiratory chain complex IV deficiency, appear sufficient to induce pain hypersensitivity, by ADP activation of P2Y1 receptors.


Subject(s)
Electron Transport Complex IV/genetics , Hypersensitivity/pathology , Mitochondria/metabolism , Mutation/genetics , Neurons, Afferent/pathology , Pain/pathology , Receptors, Purinergic P2Y1/metabolism , Adenosine Diphosphate/metabolism , Adenosine Monophosphate/metabolism , Alkyl and Aryl Transferases/metabolism , Animals , Behavior, Animal , Calcium/metabolism , Cells, Cultured , Electron Transport Complex IV/metabolism , Fluorescence , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Hypersensitivity/complications , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/drug effects , Neurons, Afferent/drug effects , Neurons, Afferent/metabolism , Nociception/drug effects , Pain/complications , Phenotype , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Kainic Acid/metabolism , Spinal Cord/pathology , Synapses/drug effects , Synapses/metabolism
6.
Sci Rep ; 7: 44169, 2017 03 10.
Article in English | MEDLINE | ID: mdl-28281561

ABSTRACT

Endometriosis is an incurable gynecological disorder characterized by debilitating pain and the establishment of innervated endometriosis lesions outside the uterus. In a preclinical mouse model of endometriosis we demonstrated overexpression of the PGE2-signaling pathway (including COX-2, EP2, EP4) in endometriosis lesions, dorsal root ganglia (DRG), spinal cord, thalamus and forebrain. TRPV1, a PGE2-regulated channel in nociceptive neurons was also increased in the DRG. These findings support the concept that an amplification process occurs along the pain neuroaxis in endometriosis. We then tested TRPV1, EP2, and EP4 receptor antagonists: The EP2 antagonist was the most efficient analgesic, reducing primary hyperalgesia by 80% and secondary hyperalgesia by 40%. In this study we demonstrate reversible peripheral and central hyperalgesia in mice with induced endometriosis.


Subject(s)
Endometriosis/drug therapy , Hyperalgesia/drug therapy , Indoles/pharmacology , Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors , Animals , Disease Models, Animal , Endometriosis/metabolism , Endometriosis/pathology , Female , Hyperalgesia/metabolism , Hyperalgesia/pathology , Mice
7.
Neuropharmacology ; 79: 136-51, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24269608

ABSTRACT

Effective relief from chronic hypersensitive pain states remains an unmet need. Here we report the discovery that the TRPM8 ion channel, co-operating with the 5-HT(1B) receptor (5-HT(1B)R) in a subset of sensory afferents, exerts an influence at the spinal cord level to suppress central hypersensitivity in pain processing throughout the central nervous system. Using cell line models, ex vivo rat neural tissue and in vivo pain models, we assessed functional Ca(2+) fluorometric responses, protein:protein interactions, immuno-localisation and reflex pain behaviours, with pharmacological and molecular interventions. We report 5-HT(1B)R expression in many TRPM8-containing afferents and direct interaction of these proteins in a novel multi-protein signalling complex, which includes phospholipase D1 (PLD1). We provide evidence that the 5-HT(1B)R activates PLD1 to subsequently activate PIP 5-kinase and generate PIP2, an allosteric enhancer of TRPM8, achieving a several-fold increase in potency of TRPM8 activation. The enhanced activation responses of synaptoneurosomes prepared from spinal cord and cortical regions of animals with a chronic inflammatory pain state are inhibited by TRPM8 activators that were applied in vivo topically to the skin, an effect potentiated by co-administered 5-HT(1B)R agonists and attenuated by 5-HT(1B)R antagonists, while 5-HT(1B)R agents alone had no detectable effect. Corresponding results are seen when assessing reflex behaviours in inflammatory and neuropathic pain models. Control experiments with alternative receptor/TRP channel combinations reveal no such synergy. Identification of this novel receptor/effector/channel complex and its impact on nociceptive processing give new insights into possible strategies for enhanced analgesia in chronic pain.


Subject(s)
Pain/metabolism , Phospholipase D/metabolism , Receptor, Serotonin, 5-HT1B/metabolism , TRPM Cation Channels/metabolism , Animals , Calcium Channels/metabolism , Cells, Cultured , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiopathology , HEK293 Cells , Humans , Male , Nerve Tissue Proteins/metabolism , Neuralgia/drug therapy , Neuralgia/physiopathology , Pain/drug therapy , Rats , Rats, Wistar , Serotonin 5-HT1 Receptor Agonists/pharmacology , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Spinal Cord/drug effects , Spinal Cord/physiopathology , TRPA1 Cation Channel , TRPV Cation Channels/metabolism , Transient Receptor Potential Channels/metabolism
8.
Int J Biochem Cell Biol ; 45(11): 2706-12, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23932932

ABSTRACT

Adverse events at critical stages of development can lead to lasting dysfunction in the central nervous system (CNS). To seek potential underlying changes in synaptic function, we used a newly developed protocol to measure alterations in receptor-mediated Ca(2+) fluorescence responses of synaptoneurosomes, freshly isolated from selected regions of the CNS concerned with emotionality and pain processing. We compared adult male controls and offspring of rats exposed to social stress in late pregnancy (prenatal stress, PS), which showed programmed behavioural changes indicating anxiety, anhedonia and pain hypersensitivity. We found corresponding increases, in PS rats compared with normal controls, in responsiveness of synaptoneurosomes from frontal cortex to a glutamate receptor (GluR) agonist, and from spinal cord to activators of nociceptive afferents. Through a combined pharmacological and biochemical strategy, we found evidence for a role of phospholipase D1 (PLD1)-mediated signalling, that may involve 5-HT2A receptor (5-HT2AR) activation, at both levels of the nervous system. These changes might participate in underpinning the enduring alterations in behaviour induced by PS.


Subject(s)
Behavior, Animal , Pain/enzymology , Pain/psychology , Phospholipase D/metabolism , Stress, Psychological/enzymology , Synapses/enzymology , Animals , Cerebral Cortex/pathology , Female , Homocysteine/analogs & derivatives , Homocysteine/pharmacology , Hypersensitivity/complications , Hypersensitivity/enzymology , Hypersensitivity/pathology , Male , Neurons/drug effects , Neurons/enzymology , Neurons/pathology , Nociception/drug effects , Pain/complications , Pain/pathology , Pregnancy , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT2A/metabolism , Spinal Cord/drug effects , Spinal Cord/enzymology , Spinal Cord/pathology , Stress, Psychological/complications , Stress, Psychological/pathology , Synapses/drug effects , Synapses/pathology
9.
PLoS One ; 8(1): e54197, 2013.
Article in English | MEDLINE | ID: mdl-23349824

ABSTRACT

Functional magnetic resonance imaging (fMRI) is a powerful method for exploring emotional and cognitive brain responses in humans. However rodent fMRI has not previously been applied to the analysis of learned behaviour in awake animals, limiting its use as a translational tool. Here we have developed a novel paradigm for studying brain activation in awake rats responding to conditioned stimuli using fMRI. Using this method we show activation of the amygdala and related fear circuitry in response to a fear-conditioned stimulus and demonstrate that the magnitude of fear circuitry activation is increased following early life stress, a rodent model of affective disorders. This technique provides a new translatable method for testing environmental, genetic and pharmacological manipulations on emotional and cognitive processes in awake rodent models.


Subject(s)
Amygdala/physiology , Fear/physiology , Magnetic Resonance Imaging/methods , Wakefulness/physiology , Animals , Brain Mapping , Conditioning, Classical , Fear/psychology , Humans , Male , Neural Pathways/physiology , Rats
10.
Cell Signal ; 25(4): 814-21, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23314176

ABSTRACT

The 5-HT2A receptor (5-HT2AR) is implicated in psychotropic changes within the central nervous system (CNS). A number of polymorphisms have been reported in the 5-HT2AR gene; one of these results in a non-synonymous change, H452Y, in the carboxy-terminal tail of the receptor protein. The minor allele (9% occurrence) has been statistically associated with CNS dysfunction such as impaired memory processing and resistance to neuroleptic treatment in schizophrenic patients. We investigated the impact of H452Y mutation of the 5-HT2AR expressed in COS7 cells on distinctly coupled intracellular signalling pathways from the receptor, focusing on the heterotrimeric G protein-independent phospholipase D (PLD) pathway, compared to the conventional Gq/11-linked phospholipase C (PLC) pathway. The H452Y mutation selectively attenuated PLD signalling, which as in the wild-type receptor, was mediated by a molecular complex involving PLD1 docked to the receptor's carboxy-terminal tail domain. Co-immunoprecipitation and GST-fusion protein experiments revealed that the H452Y mutation selectively reduced PLD1 binding to the receptor. Experiments with blocking peptides to mimic short sections of the 5-HT2AR tail sequence revealed that the peptide spanning residue 452 strongly reduced PLD but not PLC responses of the receptor. Similar observations were made when assessing both PLD responses and PLD-dependent cellular proliferation elicited by activation of 5-HT2ARs natively expressed in MCF-7 cells. Overall these findings indicate that the H452Y polymorphic variant of the 5-HT2AR displays selective disruption of its PLD signalling pathway. This may potentially play a role in the CNS dysfunction associated with the H452Y allele of the 5-HT2AR.


Subject(s)
Phospholipase D/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Animals , COS Cells , Chlorocebus aethiops , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , MCF-7 Cells , Phospholipase D/antagonists & inhibitors , Polymorphism, Single Nucleotide , Protein Isoforms/metabolism , Receptor, Serotonin, 5-HT2A/chemistry , Receptor, Serotonin, 5-HT2A/genetics , Serotonin 5-HT2 Receptor Agonists/pharmacology , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Signal Transduction/drug effects , Transfection , Type C Phospholipases/metabolism
11.
Neurosci Res ; 74(3-4): 230-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23131427

ABSTRACT

Many clinical cases of chronic pain exhibit both neuropathic and inflammatory components. In contrast, most animal models of chronic pain focus on one type of injury alone. Here we present a novel combined model of both neuropathic and inflammatory pain and characterise its distinctive properties. This combined model of chronic constriction injury (CCI) and intraplantar Complete Freund's Adjuvant (CFA) injection results in enhanced mechanical allodynia, thermal hyperalgesia, a static weight bearing deficit, and notably pronounced spontaneous foot lifting (SFL) behaviour (which under our conditions was not seen in either individual model and may reflect ongoing/spontaneous pain). Dorsal root ganglion (DRG) expression of Activating Transcription Factor-3 (ATF-3), a marker of axonal injury, was no greater in the combined model than CCI alone. Initial pharmacological characterisation of the new model showed that the SFL was reversed by gabapentin or diclofenac, typical analgesics for neuropathic or inflammatory pain respectively, but not by mexiletine, a Na(+) channel blocker effective in both neuropathic and inflammatory pain models. Static weight bearing deficit was moderately reduced by gabapentin, whereas only diclofenac reversed mechanical allodynia. This novel animal model of chronic pain may prove a useful test-bed for further analysing the pharmacological susceptibility of complicated clinical pain states.


Subject(s)
Chronic Pain , Disease Models, Animal , Inflammation , Neuralgia , Activating Transcription Factor 3/analysis , Activating Transcription Factor 3/biosynthesis , Adjuvants, Immunologic/toxicity , Animals , Behavior, Animal/physiology , Chronic Pain/metabolism , Chronic Pain/physiopathology , Freund's Adjuvant/toxicity , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Immunohistochemistry , Inflammation/metabolism , Inflammation/physiopathology , Male , Neuralgia/metabolism , Neuralgia/physiopathology , Rats , Rats, Sprague-Dawley , Sciatic Nerve/injuries
13.
Biochem J ; 436(3): 651-60, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21410433

ABSTRACT

The 5-HT2AR (5-hydroxytryptamine-2A receptor) is a GPCR (G-protein-coupled receptor) that is implicated in the actions of hallucinogens and represents a major target of atypical antipsychotic agents. In addition to its classical signalling though PLC (phospholipase C), the receptor can activate several other pathways, including ARF (ADP-ribosylation factor)-dependent activation of PLD (phospholipase D), which appears to be achieved through a mechanism independent of heterotrimeric G-proteins. In the present study we show that wild-type and inactive constructs of PLD1 (but not PLD2) respectively facilitate and inhibit ARF-dependent PLD signalling by the 5-HT2AR. Furthermore we demonstrate that PLD1 specifically co-immunoprecipitates with the receptor and binds to a distal site in GST (glutathione transferase) fusion protein constructs of its C-terminal tail which is distinct from the ARF-interaction site, thereby suggesting the existence of a functional ARF-PLD signalling complex directly associated with this receptor. This reveals the spatial co-ordination of an important GPCR, transducer and effector into a physical complex that is likely to reinforce the impact of receptor activation on a heterotrimeric G-protein-independent signalling pathway. Signalling of this receptor through such non-canonical pathways may be important to its role in particular disorders.


Subject(s)
ADP-Ribosylation Factors/metabolism , Phospholipase D/physiology , Receptor, Serotonin, 5-HT2A/physiology , Signal Transduction/physiology , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Phospholipase D/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...