Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
3.
Eur J Pain ; 26(9): 1967-1978, 2022 10.
Article in English | MEDLINE | ID: mdl-35900227

ABSTRACT

BACKGROUND: Transient receptor potential vanilloid-1 (TRPV1), activated by heat, acidic pH, endogenous vanilloids and capsaicin, is essential for thermal hyperalgesia. Under inflammatory conditions, phosphorylation of TRPV1 by protein kinase C (PKC) can sensitize the channel and decrease the activation threshold. Src kinase also phosphorylates TRPV1, promoting channel trafficking to the plasma membrane. These post-translational modifications are important for several chronic pain conditions. This study presents a previously undescribed relationship between Src and PKC phosphorylation of TRPV1, influencing the thermal hypersensitivity associated with TRPV1 activation. METHODS: We assessed TRPV1 channel activity using intracellular calcium imaging and patch-clamp electrophysiology in mouse dorsal root ganglion cultures. Additionally, we used behavioural experiments to evaluate plantar thermal sensitivity following intraplantar injections of activators of known modulators of TRPV1 with and without an Src antagonist. RESULTS: Using calcium imaging and patch-clamp techniques, we demonstrated that pharmacological inhibition of Src kinase or mutation of the Src phosphorylation site on TRPV1 prevented PKC but not PKA-mediated sensitization of TRPV1 in vitro. We found that intraplantar injection of the PKC activator phorbol 12-myristate 13-acetate (PMA) or bradykinin produces thermal hypersensitivity that can be attenuated by pharmacological inhibition of Src. Additionally, complete Freund's Adjuvant (CFA)-induced inflammatory hypersensitivity could also be attenuated by local Src kinase inhibition. CONCLUSIONS: Our data demonstrate that Src phosphorylation is critical for PKC-mediated sensitization of TRPV1. Further, in a model of inflammatory pain, CFA, Src kinase inhibition could reduce thermal hypersensitivity. Targeting of Src kinase may have analgesic benefits in inflammatory pain conditions. SIGNIFICANCE: Src kinase-mediated phosphorylation of TRPV1 is a critical regulator of the PKC-induced sensitization induced by multiple inflammatory mediators. This suggest a new regulatory mechanism governing TRPV1 function and a potential therapeutic target for inflammatory type pain, including cancer pain where Src antagonists are currently utilized.


Subject(s)
Chronic Pain , Protein Kinase C , TRPV Cation Channels , src-Family Kinases , Animals , Calcium/metabolism , Capsaicin/pharmacology , Chronic Pain/metabolism , Freund's Adjuvant/adverse effects , Ganglia, Spinal/metabolism , Hyperalgesia , Mice , Phosphorylation , Protein Kinase C/metabolism , TRPV Cation Channels/metabolism , src-Family Kinases/metabolism
4.
Big Data ; 9(4): 303-321, 2021 08.
Article in English | MEDLINE | ID: mdl-34271836

ABSTRACT

In this study, we set up a scalable framework for large-scale data processing and analytics using the big data framework. The popular classification methods are implemented, tuned, and evaluated by using intrusion datasets. The objective is to select the best classifier after optimizing the hyper-parameters. We observed that the decision tree (DT) approach outperforms compared with other methods in terms of classification accuracy, fast training time, and improved average prediction rate. Therefore, it is selected as a base classifier in our proposed ensemble approach to study class imbalance. As the intrusion datasets are imbalanced, most of the classification techniques are biased toward the majority class. The misclassification rate is more in the case of the minority class. An ensemble-based method is proposed by using K-Means, RUSBoost, and DT approaches to mitigate the class imbalance problem; empirically investigate the impact of class imbalance on classification approaches' performance; and compare the result by using popular performance metrics such as Balanced Accuracy, Matthews Correlation Coefficient, and F-Measure, which are more suitable for the assessment of imbalanced datasets.


Subject(s)
Algorithms , Big Data
5.
Anesth Analg ; 128(6): e84-e87, 2019 06.
Article in English | MEDLINE | ID: mdl-31094778

ABSTRACT

Recent findings from a phase II clinical trial showed analgesic effects of an angiotensin II type-2 receptor (AT2R) antagonist in postherpetic neuralgia patients. This study aimed to investigate whether AT2R antagonism could provide effective analgesia in voluntary measures of unevoked/ongoing pain-like behaviors in mice with experimental neuropathy. Mice were subjected to spared nerve injury to induce neuropathy and tested in 2 operant behavioral tests to measure ongoing mechanical and cold pain hypersensitivities. Systemic administration of an AT2R antagonist provided effective analgesia in these behavioral measures of mechanical and cold pain in spared nerve injury mice, suggesting its effectiveness in neuropathic pain.


Subject(s)
Angiotensin II Type 2 Receptor Blockers/administration & dosage , Cold Temperature , Ganglia, Spinal/drug effects , Hyperalgesia/drug therapy , Neuralgia/drug therapy , Analgesia , Angiotensin II/metabolism , Animals , Behavior, Animal , Female , Gait , Imidazoles/administration & dosage , Male , Mice , Mice, Inbred C57BL , Pain Management , Pyridines/administration & dosage , Receptor, Angiotensin, Type 2/metabolism
6.
Pain ; 160(3): 702-711, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30507785

ABSTRACT

The complement system significantly contributes to the development of inflammatory and neuropathic pain, but the underlying mechanisms are poorly understood. Recently, we identified the signaling pathway responsible for thermal hypersensitivity induced by the complement system component C5a. Here, we examine the mechanisms of another important action of C5a, induction of mechanical hypersensitivity. We found that intraplantar injection of C5a produced a dose-dependent mechanical sensitization and that this effect was blocked by chemogenetic ablation of macrophages in both male and female mice. Knockout of TRPV1 or pretreatment with the TRPV1 antagonists, AMG9810 or 5'-iodoresiniferatoxin (5'-IRTX), significantly reduced C5a-induced mechanical sensitization. Notably, local administration of 5'-IRTX 90 minutes after C5a injection resulted in a slow, but complete, reversal of mechanical sensitization, indicating that TRPV1 activity was required for maintaining C5a-induced mechanical hypersensitivity. This slow reversal suggests that neurogenic inflammation and neuropeptide release may be involved. Indeed, pretreatment with a calcitonin gene-related peptide (CGRP) receptor antagonist (but not an antagonist of the neurokinin 1 receptor) prevented C5a-induced mechanical sensitization. Furthermore, intraplantar injection of CGRP produced significant mechanical sensitization in both wild-type and TRPV1 knockout mice. Taken together, these findings suggest that C5a produces mechanical sensitization by initiating macrophage-to-sensory-neuron signaling cascade that involves activation of TRPV1 and CGRP receptor as critical steps in this process.


Subject(s)
Complement C5a/toxicity , Hyperalgesia/chemically induced , Hyperalgesia/pathology , Macrophages/physiology , Receptors, Calcitonin Gene-Related Peptide/metabolism , TRPV Cation Channels/metabolism , Acrylamides/pharmacology , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Dipeptides/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Nerve Growth Factor/metabolism , Pain Measurement , Piperidines/pharmacology , Quinazolines/pharmacology , Quinuclidines/pharmacology , Receptors, Calcitonin Gene-Related Peptide/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , TRPA1 Cation Channel/genetics , TRPA1 Cation Channel/metabolism , TRPV Cation Channels/genetics
7.
Proc Natl Acad Sci U S A ; 115(34): E8057-E8066, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30082378

ABSTRACT

Peripheral nerve damage initiates a complex series of structural and cellular processes that culminate in chronic neuropathic pain. The recent success of a type 2 angiotensin II (Ang II) receptor (AT2R) antagonist in a phase II clinical trial for the treatment of postherpetic neuralgia suggests angiotensin signaling is involved in neuropathic pain. However, transcriptome analysis indicates a lack of AT2R gene (Agtr2) expression in human and rodent sensory ganglia, raising questions regarding the tissue/cell target underlying the analgesic effect of AT2R antagonism. We show that selective antagonism of AT2R attenuates neuropathic but not inflammatory mechanical and cold pain hypersensitivity behaviors in mice. Agtr2-expressing macrophages (MΦs) constitute the predominant immune cells that infiltrate the site of nerve injury. Interestingly, neuropathic mechanical and cold pain hypersensitivity can be attenuated by chemogenetic depletion of peripheral MΦs and AT2R-null hematopoietic cell transplantation. Our study identifies AT2R on peripheral MΦs as a critical trigger for pain sensitization at the site of nerve injury, and therefore proposes a translatable peripheral mechanism underlying chronic neuropathic pain.


Subject(s)
Chronic Pain/metabolism , Macrophages/metabolism , Neuralgia/metabolism , Receptor, Angiotensin, Type 2/metabolism , Allografts , Animals , Chronic Pain/genetics , Chronic Pain/pathology , Hematopoietic Stem Cell Transplantation , Macrophages/pathology , Mice , Neuralgia/genetics , Neuralgia/pathology , Receptor, Angiotensin, Type 2/genetics
8.
J Neurosci ; 38(32): 7032-7057, 2018 08 08.
Article in English | MEDLINE | ID: mdl-29976627

ABSTRACT

Injury, inflammation, and nerve damage initiate a wide variety of cellular and molecular processes that culminate in hyperexcitation of sensory nerves, which underlies chronic inflammatory and neuropathic pain. Using behavioral readouts of pain hypersensitivity induced by angiotensin II (Ang II) injection into mouse hindpaws, our study shows that activation of the type 2 Ang II receptor (AT2R) and the cell-damage-sensing ion channel TRPA1 are required for peripheral mechanical pain sensitization induced by Ang II in male and female mice. However, we show that AT2R is not expressed in mouse and human dorsal root ganglia (DRG) sensory neurons. Instead, expression/activation of AT2R on peripheral/skin macrophages (MΦs) constitutes a critical trigger of mouse and human DRG sensory neuron excitation. Ang II-induced peripheral mechanical pain hypersensitivity can be attenuated by chemogenetic depletion of peripheral MΦs. Furthermore, AT2R activation in MΦs triggers production of reactive oxygen/nitrogen species, which trans-activate TRPA1 on mouse and human DRG sensory neurons via cysteine modification of the channel. Our study thus identifies a translatable immune cell-to-sensory neuron signaling crosstalk underlying peripheral nociceptor sensitization. This form of cell-to-cell signaling represents a critical peripheral mechanism for chronic pain and thus identifies multiple druggable analgesic targets.SIGNIFICANCE STATEMENT Pain is a widespread health problem that is undermanaged by currently available analgesics. Findings from a recent clinical trial on a type II angiotensin II receptor (AT2R) antagonist showed effective analgesia for neuropathic pain. AT2R antagonists have been shown to reduce neuropathy-, inflammation- and bone cancer-associated pain in rodents. We report that activation of AT2R in macrophages (MΦs) that infiltrate the site of injury, but not in sensory neurons, triggers an intercellular redox communication with sensory neurons via activation of the cell damage/pain-sensing ion channel TRPA1. This MΦ-to-sensory neuron crosstalk results in peripheral pain sensitization. Our findings provide an evidence-based mechanism underlying the analgesic action of AT2R antagonists, which could accelerate the development of efficacious non-opioid analgesic drugs for multiple pain conditions.


Subject(s)
Angiotensin II/physiology , Hyperalgesia/physiopathology , Macrophages, Peritoneal/metabolism , Neuralgia/physiopathology , Receptor, Angiotensin, Type 2/physiology , Sensory Receptor Cells/physiology , TRPA1 Cation Channel/physiology , Angiotensin II/toxicity , Angiotensin Receptor Antagonists/pharmacology , Animals , Cell Communication/physiology , Cells, Cultured , Female , Ganglia, Spinal/cytology , Genes, Reporter , Humans , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Imidazoles/pharmacology , Macrophage Activation , Macrophages, Peritoneal/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuralgia/drug therapy , Neutrophil Activation , Oxidation-Reduction , Pyridines/pharmacology , Receptor, Angiotensin, Type 2/genetics , Sensory Receptor Cells/chemistry , Skin/cytology , TRPA1 Cation Channel/deficiency , Tacrolimus/analogs & derivatives , Tacrolimus/pharmacology
9.
Front Behav Neurosci ; 12: 124, 2018.
Article in English | MEDLINE | ID: mdl-30002622

ABSTRACT

Burrowing, or the removal of material from an enclosed tube, is emerging as a prominent means of testing changes in a voluntary behavior in rodent models of various pain states. Here, we report no significant differences between male and female mice in terms of burrowing performance, in a substantially shorter time frame than previous reports. We found that the color of the burrow tube affects the variability of burrowing performance when tested in a lit room, suggesting that light aversion is at least a partial driver of this behavior. Spared nerve injury (SNI; as a model of neuropathy) impairs burrowing performance and correlates with enhanced mechanical sensitivity as assessed by von Frey filaments, as well as being pharmacologically reversed by an analgesic, gabapentin. Loss of the SNI-induced burrowing deficit was observed with daily testing post-surgery, but not when the testing interval was increased to 5 days, suggesting a confounding effect of daily repeat testing in this paradigm. Intraplantar complete Freund's adjuvant (as a model of inflammatory pain) and systemic nitroglycerin (as a model of migraine-like symptoms) administration did not induce any burrowing deficit, indicating that assessment of burrowing behavior may not be universally suitable for the detection of behavioral changes across all rodent pain models.

10.
Front Cell Neurosci ; 12: 38, 2018.
Article in English | MEDLINE | ID: mdl-29497363

ABSTRACT

Bone metastasis in breast, prostate and lung cancers often leads to chronic pain, which is poorly managed by existing analgesics. The neurobiological mechanisms that underlie chronic pain associated with bone-metastasized cancers are not well understood, but sensitization of peripheral nociceptors by tumor microenvironment factors has been demonstrated to be important. Parathyroid hormone-related peptide (PTHrP) is highly expressed in bone-metastasized breast and prostate cancers, and is critical to growth and proliferation of these tumors in the bone tumor microenvironment. Previous studies have suggested that PTHrP could sensitize nociceptive sensory neurons, resulting in peripheral pain hypersensitivity. In this study, we found that PTHrP induces both heat and mechanical hypersensitivity, that are dependent on the pain-transducing transient receptor potential channel family vanilloid, member-1 (TRPV1), but not the mechano-transducing TRPV4 and TRPA1 ion channels. Functional ratiometric Ca2+ imaging and voltage-clamp electrophysiological analysis of cultured mouse DRG neurons show significant potentiation of TRPV1, but not TRPA1 or TRPV4 channel activation by PTHrP. Interestingly, PTHrP exposure led to the slow and sustained activation of TRPV1, in the absence of any exogenous channel agonist, and is dependent on the expression of the type-1 parathyroid hormone receptor (PTH1), as well as on downstream phosphorylation of the channel by protein kinase C (PKC). Accordingly, local administration of specific small-molecule antagonists of TRPV1 to mouse hindpaws after the development of PTHrP-induced mechanical hypersensitivity led to its significant attenuation. Collectively, our findings suggest that PTHrP/PTH1-mediated flow activation of TRPV1 channel contributes at least in part to the development and maintenance of peripheral mechanical pain hypersensitivity, and could therefore constitute a mechanism for nociceptor sensitization in the context of metastatic bone cancer pain.

11.
Neuron ; 97(5): 1094-1109.e9, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29429936

ABSTRACT

Despite the central role PSD-95 plays in anchoring postsynaptic AMPARs, how PSD-95 itself is tethered to postsynaptic sites is not well understood. Here we show that the F-actin binding protein α-actinin binds to the very N terminus of PSD-95. Knockdown (KD) of α-actinin phenocopies KD of PSD-95. Mutating lysine at position 10 or lysine at position 11 of PSD-95 to glutamate, or glutamate at position 53 or glutamate and aspartate at positions 213 and 217 of α-actinin, respectively, to lysine impairs, in parallel, PSD-95 binding to α-actinin and postsynaptic localization of PSD-95 and AMPARs. These experiments identify α-actinin as a critical PSD-95 anchor tethering the AMPAR-PSD-95 complex to postsynaptic sites.


Subject(s)
Actinin/metabolism , Disks Large Homolog 4 Protein/metabolism , Excitatory Postsynaptic Potentials/physiology , Hippocampus/metabolism , Actinin/chemistry , Actinin/genetics , Amino Acid Sequence , Animals , Cells, Cultured , Disks Large Homolog 4 Protein/chemistry , Disks Large Homolog 4 Protein/genetics , Female , HEK293 Cells , Humans , Male , Protein Structure, Secondary , Rats
12.
Neuropharmacology ; 130: 18-29, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29191755

ABSTRACT

The urgent need for more effective analgesic treatment options has prompted a re-evaluation of the behavioral tests used to assess pain in pre-clinical research, with an emphasis on inclusion of more voluntary, un-evoked behavioral assessments of pain. In order to validate voluntary gait analysis and a voluntary mechanical conflict-avoidance assay, we tested mouse models of neuropathy (spared nerve injury) and inflammation (complete Freund's adjuvant) alongside reflexive measures of mechanical and thermal hypersensitivity. To establish whether the observed changes in behavioral responses were pain-related, known analgesics (buprenorphine, gabapentin, carprofen) were also administered. Spared nerve injury persistently altered several gait indices, whereas complete Freund's adjuvant caused only transient changes. Furthermore, known analgesics could not reverse these gait changes, despite demonstrating their previously established efficacy in reflexive measures of mechanical and thermal hypersensitivity. In contrast, the mechanical conflict-avoidance assay demonstrated aversion in mice with neuropathy and inflammation-induced hypersensitivity, which could both be reversed by analgesics. We conclude that voluntary gait changes in rodent neuropathic and inflammatory pain models are not necessarily indicative of pain-related adaptations. On the other hand, mechanical conflict-avoidance represents a valid operant assay for quantifying pain-related behaviors in mice that can be reversed by known analgesics.


Subject(s)
Analgesics/pharmacology , Gait/drug effects , Neuralgia/drug therapy , Neuralgia/physiopathology , Animals , Buprenorphine/pharmacology , Carbazoles/pharmacology , Disease Models, Animal , Female , Freund's Adjuvant/pharmacology , Gabapentin/pharmacology , Gait/physiology , Inflammation/drug therapy , Inflammation/physiopathology , Male , Mice , Mice, Inbred C57BL , Pain Measurement/drug effects , Pain Measurement/methods
13.
Neurobiol Pain ; 2: 1-12, 2017.
Article in English | MEDLINE | ID: mdl-29075674

ABSTRACT

It has been suggested that the lack of rodent behavioral assays that represent the complexities of human pain contributes to the poor translational record of basic pain research findings. Clinically, chronic pain interferes with patient mobility and physical/social activities, and increases anxiety symptoms, in turn negatively impacting quality of life. To determine whether these behaviors are similarly influenced by putative pain manipulations in rodents, we systematically evaluated wheel running, locomotion, gait, social interaction, and anxiety-like behavior in models of inflammation and nerve injury in adult C57BL6/J male mice. We demonstrate that inflammation and nerve injury differentially affect voluntary behaviors while mice are hypersensitive to mechanical stimuli. Bilateral Complete Freund's Adjuvant (CFA)-induced inflammation transiently suppressed wheel running and locomotion and also induced gait deficits. In contrast, spared nerve injury (SNI) altered gait and impaired gross motor coordination. SNI-induced gait changes were not reversed by the analgesic PD123319, an angiotensin II type 2 receptor antagonist, and are therefore likely to be motor-related rather than pain-related. Neither CFA nor SNI significantly altered social interaction or elicited general anxiety-like behavior. Our findings suggest that in contrast to humans, mobility and physical/social activities are minimally altered, if at all, in mice following inflammation or nerve injury.

14.
Curr Pain Headache Rep ; 21(6): 28, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28432601

ABSTRACT

PURPOSE OF REVIEW: Our goal is to examine the processes-both central and peripheral-that underlie the development of peripherally-induced neuropathic pain (pNP) and to highlight recent evidence for mechanisms contributing to its maintenance. While many pNP conditions are initiated by damage to the peripheral nervous system (PNS), their persistence appears to rely on maladaptive processes within the central nervous system (CNS). The potential existence of an autonomous pain-generating mechanism in the CNS creates significant implications for the development of new neuropathic pain treatments; thus, work towards its resolution is crucial. Here, we seek to identify evidence for PNS and CNS independently generating neuropathic pain signals. RECENT FINDINGS: Recent preclinical studies in pNP support and provide key details concerning the role of multiple mechanisms leading to fiber hyperexcitability and sustained electrical discharge to the CNS. In studies regarding central mechanisms, new preclinical evidence includes the mapping of novel inhibitory circuitry and identification of the molecular basis of microglia-neuron crosstalk. Recent clinical evidence demonstrates the essential role of peripheral mechanisms, mostly via studies that block the initially damaged peripheral circuitry. Clinical central mechanism studies use imaging to identify potentially self-sustaining infra-slow CNS oscillatory activity that may be unique to pNP patients. While new preclinical evidence supports and expands upon the key role of central mechanisms in neuropathic pain, clinical evidence for an autonomous central mechanism remains relatively limited. Recent findings from both preclinical and clinical studies recapitulate the critical contribution of peripheral input to maintenance of neuropathic pain. Further clinical investigations on the possibility of standalone central contributions to pNP may be assisted by a reconsideration of the agreed terms or criteria for diagnosing the presence of central sensitization in humans.


Subject(s)
Central Nervous System/physiopathology , Neuralgia/etiology , Peripheral Nervous System/physiopathology , Humans , Neuralgia/physiopathology , Neurons/physiology , Peripheral Nervous System/injuries
15.
Pharmaceuticals (Basel) ; 9(4)2016 Nov 14.
Article in English | MEDLINE | ID: mdl-27854251

ABSTRACT

Specialized receptors belonging to the transient receptor potential (TRP) family of ligand-gated ion channels constitute the critical detectors and transducers of pain-causing stimuli. Nociceptive TRP channels are predominantly expressed by distinct subsets of sensory neurons of the peripheral nervous system. Several of these TRP channels are also expressed in neurons of the central nervous system, and in non-neuronal cells that communicate with sensory nerves. Nociceptive TRPs are activated by specific physico-chemical stimuli to provide the excitatory trigger in neurons. In addition, decades of research has identified a large number of immune and neuromodulators as mediators of nociceptive TRP channel activation during injury, inflammatory and other pathological conditions. These findings have led to aggressive targeting of TRP channels for the development of new-generation analgesics. This review summarizes the complex activation and/or modulation of nociceptive TRP channels under pathophysiological conditions, and how these changes underlie acute and chronic pain conditions. Furthermore, development of small-molecule antagonists for several TRP channels as analgesics, and the positive and negative outcomes of these drugs in clinical trials are discussed. Understanding the diverse functional and modulatory properties of nociceptive TRP channels is critical to function-based drug targeting for the development of evidence-based and efficacious new generation analgesics.

16.
J Neurosci ; 36(18): 5055-70, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27147658

ABSTRACT

UNLABELLED: The complement cascade is a principal component of innate immunity. Recent studies have underscored the importance of C5a and other components of the complement system in inflammatory and neuropathic pain, although the underlying mechanisms are largely unknown. In particular, it is unclear how the complement system communicates with nociceptors and which ion channels and receptors are involved. Here we demonstrate that inflammatory thermal and mechanical hyperalgesia induced by complete Freund's adjuvant was accompanied by C5a upregulation and was markedly reduced by C5a receptor (C5aR1) knock-out or treatment with the C5aR1 antagonist PMX53. Direct administration of C5a into the mouse hindpaw produced strong thermal hyperalgesia, an effect that was absent in TRPV1 knock-out mice, and was blocked by the TRPV1 antagonist AMG9810. Immunohistochemistry of mouse plantar skin showed prominent expression of C5aR1 in macrophages. Additionally, C5a evoked strong Ca(2+) mobilization in macrophages. Macrophage depletion in transgenic macrophage Fas-induced apoptosis mice abolished C5a-dependent thermal hyperalgesia. Examination of inflammatory mediators following C5a injection revealed a rapid upregulation of NGF, a mediator known to sensitize TRPV1. Preinjection of an NGF-neutralizing antibody or Trk inhibitor GNF-5837 prevented C5a-induced thermal hyperalgesia. Notably, NGF-induced thermal hyperalgesia was unaffected by macrophage depletion. Collectively, these results suggest that complement fragment C5a induces thermal hyperalgesia by triggering macrophage-dependent signaling that involves mobilization of NGF and NGF-dependent sensitization of TRPV1. Our findings highlight the importance of macrophage-to-neuron signaling in pain processing and identify C5a, NGF, and TRPV1 as key players in this cross-cellular communication. SIGNIFICANCE STATEMENT: This study provides mechanistic insight into how the complement system, a key component of innate immunity, regulates the development of pain hypersensitivity. We demonstrate a crucial role of the C5a receptor, C5aR1, in the development of inflammatory thermal and mechanical sensitization. By focusing on the mechanisms of C5a-induced thermal hyperalgesia, we show that this process requires recruitment of macrophages and initiation of macrophage-to-nociceptor signaling. At the molecular level, we demonstrate that this signaling depends on NGF and is mediated by the heat-sensitive nociceptive channel TRPV1. This deeper understanding of how immune cells and neurons interact to regulate pain processing is expected to facilitate mechanism-based approaches in the development of new analgesics.


Subject(s)
Complement C5a/metabolism , Hyperalgesia/physiopathology , Macrophages , Nerve Growth Factor , Nociceptors , Signal Transduction , TRPV Cation Channels , Acrylamides/pharmacology , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Communication , Complement C5a/genetics , Female , Hot Temperature , Inflammation/chemically induced , Inflammation/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nerve Growth Factor/antagonists & inhibitors , Physical Stimulation , TRPV Cation Channels/antagonists & inhibitors
17.
Pain ; 157(1): 235-246, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26397933

ABSTRACT

Obesity is associated with several pain disorders including headache. The effects of obesity on the trigeminal nociceptive system, which mediates headache, remain unknown. We used 2 complementary mouse models of obesity (high-fat diet and leptin deficiency) to examine this. We assessed capsaicin-induced nocifensive behavior and photophobia in obese and control mice. Calcium imaging was used to determine the effects of obesity on the activity of primary trigeminal afferents in vitro. We found that obese mice had a normal acute response to a facial injection of capsaicin, but they developed photophobic behavior at doses that had no effect on control mice. We observed higher calcium influx in cultured trigeminal ganglia neurons from obese mice and a higher percentage of medium to large diameter capsaicin-responsive cells. These findings demonstrate that obesity results in functional changes in the trigeminal system that may contribute to abnormal sensory processing. Our findings provide the foundation for in-depth studies to improve the understanding of the effects of obesity on the trigeminal system and may have implications for the pathophysiology of headache disorders.


Subject(s)
Neurons/physiology , Obesity/physiopathology , Pain/physiopathology , Sensation Disorders/physiopathology , Trigeminal Ganglion/physiopathology , Animals , Behavior, Animal/physiology , Capsaicin/pharmacology , Mice , Mice, Obese , Neurons/drug effects , Obesity/complications , Pain/complications , Sensation/drug effects , Sensation Disorders/complications , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiology , Trigeminal Ganglion/drug effects
18.
Neuropharmacology ; 101: 291-308, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26456351

ABSTRACT

The endogenous neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) is secreted by both neuronal and non-neuronal cells in the brain and spinal cord, in response to pathological conditions such as stroke, seizures, chronic inflammatory and neuropathic pain. PACAP has been shown to exert various neuromodulatory and neuroprotective effects. However, direct influence of PACAP on the function of intrinsically excitable ion channels that are critical to both hyperexcitation as well as cell death, remain largely unexplored. The major dendritic K(+) channel Kv4.2 is a critical regulator of neuronal excitability, back-propagating action potentials in the dendrites, and modulation of synaptic inputs. We identified, cloned and characterized the downstream signaling originating from the activation of three PACAP receptor (PAC1) isoforms that are expressed in rodent hippocampal neurons that also exhibit abundant expression of Kv4.2 protein. Activation of PAC1 by PACAP leads to phosphorylation of Kv4.2 and downregulation of channel currents, which can be attenuated by inhibition of either PKA or ERK1/2 activity. Mechanistically, this dynamic downregulation of Kv4.2 function is a consequence of reduction in the density of surface channels, without any influence on the voltage-dependence of channel activation. Interestingly, PKA-induced effects on Kv4.2 were mediated by ERK1/2 phosphorylation of the channel at two critical residues, but not by direct channel phosphorylation by PKA, suggesting a convergent phosphomodulatory signaling cascade. Altogether, our findings suggest a novel GPCR-channel signaling crosstalk between PACAP/PAC1 and Kv4.2 channel in a manner that could lead to neuronal hyperexcitability.


Subject(s)
Dendrites/drug effects , Neurons/cytology , Neurotransmitter Agents/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Shal Potassium Channels/metabolism , Animals , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation/drug effects , Hippocampus/cytology , Humans , Male , Mice , Mutagenesis/genetics , Mutation/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Rats , Rats, Sprague-Dawley , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
19.
Pain ; 156(9): 1620-1636, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25970319

ABSTRACT

The neurobiological mechanisms underlying chronic pain associated with cancers are not well understood. It has been hypothesized that factors specifically elevated in the tumor microenvironment sensitize adjacent nociceptive afferents. We show that parathyroid hormone-related peptide (PTHrP), which is found at elevated levels in the tumor microenvironment of advanced breast and prostate cancers, is a critical modulator of sensory neurons. Intraplantar injection of PTHrP led to the development of thermal and mechanical hypersensitivity in both male and female mice, which were absent in mice lacking functional transient receptor potential vanilloid-1 (TRPV1). The PTHrP treatment of cultured mouse sensory neurons enhanced action potential firing, and increased TRPV1 activation, which was dependent on protein kinase C (PKC) activity. Parathyroid hormone-related peptide induced robust potentiation of TRPV1 activation and enhancement of neuronal firing at mild acidic pH that is relevant to acidic tumor microenvironment. We also observed an increase in plasma membrane TRPV1 protein levels after exposure to PTHrP, leading to upregulation in the proportion of TRPV1-responsive neurons, which was dependent on the activity of PKC and Src kinases. Furthermore, co-injection of PKC or Src inhibitors attenuated PTHrP-induced thermal but not mechanical hypersensitivity. Altogether, our results suggest that PTHrP and mild acidic conditions could induce constitutive pathological activation of sensory neurons through upregulation of TRPV1 function and trafficking, which could serve as a mechanism for peripheral sensitization of nociceptive afferents in the tumor microenvironment.


Subject(s)
Hyperalgesia/chemically induced , Parathyroid Hormone-Related Protein/toxicity , Protein Transport/drug effects , TRPV Cation Channels/metabolism , Animals , Capsaicin/pharmacology , Cells, Cultured , Cyclic AMP/metabolism , Female , Ganglia, Spinal/cytology , Humans , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Protein Kinase C-epsilon/metabolism , Receptor, Parathyroid Hormone, Type 1/metabolism , TRPV Cation Channels/genetics , Up-Regulation/genetics
20.
Prog Mol Biol Transl Sci ; 131: 73-118, 2015.
Article in English | MEDLINE | ID: mdl-25744671

ABSTRACT

Peripheral detection of nociceptive and painful stimuli by sensory neurons involves a complex repertoire of molecular detectors and/or transducers on distinct subsets of nerve fibers. The majority of such molecular detectors/transducers belong to the transient receptor potential (TRP) family of cation channels, which comprise both specific receptors for distinct nociceptive stimuli, as well as for multiple stimuli. This chapter discusses the classification, distribution, and functional properties of individual TRP channel types that have been implicated in various nociceptive and/or painful conditions.


Subject(s)
Nociception , Pain/metabolism , Sense Organs/metabolism , Transient Receptor Potential Channels/metabolism , Animals , Humans , Models, Biological , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...