Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 220
Filter
Add more filters

Publication year range
1.
FASEB J ; 36(4): e22236, 2022 04.
Article in English | MEDLINE | ID: mdl-35218596

ABSTRACT

Lysophosphatidic acid (LPA) exerts various biological activities through six characterized G protein-coupled receptors (LPA1-6 ). While LPA-LPA1  signaling contributes toward the demyelination and retraction of C-fiber and induces neuropathic pain, the effects of LPA-LPA1  signaling on acute nociceptive pain is uncertain. This study investigated the role of LPA-LPA1  signaling in acute nociceptive pain using the formalin test. The pharmacological inhibition of the LPA-LPA1 axis significantly attenuated formalin-induced nociceptive behavior. The LPA1  mRNA was expressed in satellite glial cells (SGCs) in dorsal root ganglion (DRG) and was particularly abundant in SGCs surrounding large DRG neurons, which express neurofilament 200. Treatment with LPA1/3 receptor (LPA1/3 ) antagonist inhibited the upregulation of glial markers and inflammatory cytokines in DRG following formalin injection. The LPA1/3 antagonist also attenuated phosphorylation of extracellular signal-regulated kinase, especially in SGCs and cyclic AMP response element-binding protein in the dorsal horn following formalin injection. LPA amounts after formalin injection to the footpad were quantified by liquid chromatography/tandem mass spectrometry, and LPA levels were found to be increased in the innervated DRGs. Our results indicate that LPA produced in the innervated DRGs promotes the activation of SGCs through LPA1 , increases the sensitivity of primary neurons, and modulates pain behavior. These results facilitate our understanding of the pathology of acute nociceptive pain and demonstrate the possibility of the LPA1 on SGCs as a novel target for acute pain control.


Subject(s)
Isoxazoles/pharmacology , Lysophospholipids/metabolism , Neuroglia/drug effects , Nociceptive Pain/prevention & control , Propionates/pharmacology , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Animals , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Female , Ganglia, Spinal , Male , Mice , Mice, Inbred C57BL , Neuroglia/metabolism , Nociceptive Pain/etiology , Nociceptive Pain/metabolism , Nociceptive Pain/pathology , Phosphorylation , Signal Transduction
2.
Neural Plast ; 2023: 9303419, 2023.
Article in English | MEDLINE | ID: mdl-36910013

ABSTRACT

Referred somatic pain triggered by hyperalgesia is common in patients with inflammatory bowel disease (IBD). It was reported that sprouting of sympathetic nerve fibers into the dorsal root ganglion (DGR) and neurogenic inflammation were related to neuropathic pain, the excitability of neurons, and afferents. The purpose of the study was to explore the potential and mechanism of electroacupuncture (EA) at Zusanli (ST36) for the intervention of colon inflammation and hyperalgesia. Sprague-Dawley (SD) was randomly divided into four groups, including control, model, EA, and sham-EA. Our results showed EA treatment significantly attenuated dextran sulfate sodium- (DSS-) induced colorectal lesions and inflammatory cytokine secretion, such as TNF-α, IL-1ß, PGE2, and IL-6. EA also inhibited mechanical and thermal pain hypersensitivities of colitis rats. Importantly, EA effectively abrogated the promotion effect of DSS on ipsilateral lumbar 6 (L6) DRG sympathetic-sensory coupling, manifested as the sprouting of tyrosine hydroxylase- (TH-) positive sympathetic fibers into sensory neurons and colocalization of and calcitonin gene-related peptide (CGRP). Furthermore, EA at Zusanli (ST36) activated neurogenic inflammation, characterized by decreased expression of substance P (SP), hyaluronic acid (HA), bradykinin (BK), and prostacyclin (PGI2) in colitis rat skin tissues corresponding to the L6 DRG. Mechanically, EA treatment reduced the activation of the TRPV1/CGRP, ERK, and TLR4 signaling pathways in L6 DRG of colitis rats. Taken together, we presumed that EA treatment improved colon inflammation and hyperalgesia, potentially by suppressing the sprouting of sympathetic nerve fibers into the L6 DGR and neurogenic inflammation via deactivating the TRPV1/CGRP, ERK, and TLR4 signaling pathways.


Subject(s)
Colitis , Electroacupuncture , Neuralgia , Nociceptive Pain , Rats , Animals , Rats, Sprague-Dawley , Hyperalgesia/metabolism , Electroacupuncture/methods , Ganglia, Spinal/metabolism , Calcitonin Gene-Related Peptide/metabolism , Neurogenic Inflammation/metabolism , Toll-Like Receptor 4/metabolism , Neuralgia/metabolism , Nociceptive Pain/metabolism
3.
J Neurophysiol ; 127(2): 397-404, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34986062

ABSTRACT

Whether pinocembrin (PCN) could be used to alleviate hip fracture-induced pain is investigated in this research. Aged rats with hip fractures were treated with vehicle or 80 mg/kg/day PCN from week 3 to week 4. Then, hind paw mechanical allodynia, unweighting, warmth, and thickness were measured. The microglia and astrocytes activation and proliferation markers in the spinal dorsal horn were detected with real-time PCR and immunofluorescence staining. The relative expression of substance P and its receptor, tachykinin receptor 1 (Tacr1), was detected with enzyme-linked immunosorbent assay (ELISA) and Western blots. The antinociceptive effect of Tacr1 inhibitor LY303870 was also testified. PCN alleviated hip fracture-induced hind paw nociceptive (allodynia and unweighting) and vascular changes (warmth and thickness) in aged rats with diminished microglia and astrocytes activation and proliferation in the spinal dorsal horn. Upregulated substance P and Tacr1 were induced after hip fracture, which could be reversed by PCN treatment. Furthermore, LY303870 treatment partially reversed both spinal nociceptive sensitization and vascular changes after hip fracture. Substance P signaling contributes to the nociceptive and vascular changes observed in the hip fracture, which could be alleviated by PCN.NEW & NOTEWORTHY Substance P signaling contributes to the nociceptive and vascular changes observed in hip fracture, which could be alleviated by PCN.


Subject(s)
Aging , Flavanones/pharmacology , Hip Fractures/drug therapy , Neurokinin-1 Receptor Antagonists/pharmacology , Pain/drug therapy , Substance P/drug effects , Animals , Disease Models, Animal , Flavanones/administration & dosage , Hip Fractures/complications , Hip Fractures/metabolism , Indoles/pharmacology , Male , Neurokinin-1 Receptor Antagonists/administration & dosage , Nociceptive Pain/drug therapy , Nociceptive Pain/etiology , Nociceptive Pain/metabolism , Pain/etiology , Pain/metabolism , Piperidines/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
4.
Pflugers Arch ; 473(4): 683-695, 2021 04.
Article in English | MEDLINE | ID: mdl-33474635

ABSTRACT

The pro-resolving mechanism is a recently described endogenous process that controls inflammation. The present study evaluated components of this mechanism, including annexin 1 (ANXA1) and the formyl peptide receptor 2/ALX (FPR2/ALX) receptor, in the antihyperalgesic effect induced by electroacupuncture (EA) in an animal model of persistent peripheral inflammation. Male Swiss mice underwent intraplantar (i.pl.) injection with complete Freund's adjuvant (CFA). Mechanical hyperalgesia was assessed with von Frey monofilaments. Animals were treated with EA (2-10 Hz, ST36-SP6) or subcutaneous BML-111 injection (FPR2/ALX agonist) for 5 consecutive days. In a separate set of experiments, on the first and fifth days after CFA injection, animals received i.pl. WRW4 (FPR2/ALX antagonist) or naloxone (non-selective opioid receptor antagonist) before EA or BML-111 injection. Paw protein levels of FPR2/ALX and ANXA1 were evaluated on the second day after CFA injection by western blotting technique. EA and BML-111 reduced mechanical hyperalgesia. I.pl. naloxone or WRW4 prevented the antihyperalgesic effect induced by either EA or BML-111. EA increased ANXA1 but did not alter FPR2/ALX receptor levels in the paw. Furthermore, i.pl. pretreatment with WRW4 prevented the increase of ANXA1 levels induced by EA. This work demonstrates that the EA antihyperalgesic effect on inflammatory pain involves the ANXA1/FPR2/ALX pro-resolution pathway. This effect appears to be triggered by the activation of FPR2/ALX receptors and crosstalk communication with the opioid system.


Subject(s)
Annexin A1/metabolism , Electroacupuncture/methods , Hyperalgesia/therapy , Nociceptive Pain/therapy , Receptors, Formyl Peptide/metabolism , Receptors, Opioid/metabolism , Animals , Freund's Adjuvant/toxicity , Heptanoic Acids/pharmacology , Hyperalgesia/etiology , Hyperalgesia/metabolism , Male , Mice , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Nociception/drug effects , Nociceptive Pain/etiology , Nociceptive Pain/metabolism , Receptors, Formyl Peptide/antagonists & inhibitors , Receptors, Opioid/therapeutic use
5.
Am J Physiol Renal Physiol ; 321(5): F587-F599, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34514879

ABSTRACT

Sensitization of neuronal pathways and persistent afferent drive are major contributors to somatic and visceral pain. However, the underlying mechanisms that govern whether afferent signaling will give rise to sensitization and pain are not fully understood. In the present report, we investigated the contribution of acid-sensing ion channels (ASICs) to bladder nociception in a model of chemical cystitis induced by cyclophosphamide (CYP). We found that the administration of CYP to mice lacking ASIC3, a subunit primarily expressed in sensory neurons, generates pelvic allodynia at a time point at which only modest changes in pelvic sensitivity are apparent in wild-type mice. The differences in mechanical pelvic sensitivity between wild-type and Asic3 knockout mice treated with CYP were ascribed to sensitized bladder C nociceptors. Deletion of Asic3 from bladder sensory neurons abolished their ability to discharge action potentials in response to extracellular acidification. Collectively, the results of our study support the notion that protons and their cognate ASIC receptors are part of a mechanism that operates at the nerve terminals to control nociceptor excitability and sensitization.NEW & NOTEWORTHY Our study indicates that protons and their cognate acid-sensing ion channel receptors are part of a mechanism that operates at bladder afferent terminals to control their function and that the loss of this regulatory mechanism results in hyperactivation of nociceptive pathways and the development of pain in the setting of chemical-induced cystitis.


Subject(s)
Acid Sensing Ion Channels/metabolism , Cystitis/metabolism , Nociception , Nociceptive Pain/metabolism , Nociceptors/metabolism , Urinary Bladder/innervation , Acid Sensing Ion Channels/genetics , Action Potentials , Animals , Cyclophosphamide , Cystitis/chemically induced , Cystitis/physiopathology , Disease Models, Animal , Hydrogen-Ion Concentration , Mice, Inbred C57BL , Mice, Knockout , Nociceptive Pain/chemically induced , Nociceptive Pain/physiopathology , Urination
6.
J Neurophysiol ; 125(5): 1598-1611, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33596743

ABSTRACT

Neuroactive substances released by activated microglia contribute to hyperexcitability of spinal dorsal horn neurons in many animal models of chronic pain. An important feedback loop mechanism is via release of fractalkine (CX3CL1) from primary afferent terminals and dorsal horn neurons and binding to CX3CR1 receptors on microglial cells. We studied the involvement of fractalkine signaling in latent and manifest spinal sensitization induced by two injections of nerve growth factor (NGF) into the lumbar multifidus muscle as a model for myofascial low back pain. Single dorsal horn neurons were recorded in vivo to study their receptive fields and spontaneous activity. Under intrathecal vehicle application, the two NGF injections led to an increased proportion of neurons responding to stimulation of deep tissues (41%), to receptive field expansion into the hindlimb (15%), and to resting activity (53%). Blocking fractalkine signaling by continuous intrathecal administration of neutralizing antibodies completely prevented these signs of spinal sensitization to a similar extent as in a previous study with the microglia inhibitor minocycline. Reversely, fractalkine itself induced similar sensitization in a dose-dependent manner (for 200 ng/mL: 45% deep tissue responses, 24% receptive field expansion, and 45% resting activity) as repeated nociceptive stimulation by intramuscular NGF injections. A subsequent single NGF injection did not have an additive effect. Our data suggest that neuron-to-microglia signaling via the CX3CL1-CX3CR1 pathway is critically involved in the initiation of nonspecific, myofascial low back pain through repetitive nociceptive stimuli.NEW & NOTEWORTHY Blocking fractalkine signaling by neutralizing antibodies completely prevented spinal sensitization induced by repetitive mild nociceptive input [2 nerve growth factor (NGF) injections into the multifidus muscle] Conversely, fractalkine given intrathecally caused the same pattern of spinal sensitization as the nociceptive NGF injections. Fractalkine signaling is critically involved in sensitization of dorsal horn neurons induced by repeated nociceptive low back muscle stimulation and may hence be a potential target for the prevention of nonspecific, myofascial low back pain.


Subject(s)
CX3C Chemokine Receptor 1/metabolism , Central Nervous System Sensitization/physiology , Chemokine CX3CL1/metabolism , Low Back Pain/metabolism , Nociceptive Pain/metabolism , Posterior Horn Cells/metabolism , Signal Transduction/physiology , Animals , Antibodies, Neutralizing/pharmacology , CX3C Chemokine Receptor 1/drug effects , Central Nervous System Sensitization/drug effects , Chemokine CX3CL1/drug effects , Chemokine CX3CL1/pharmacology , Chronic Pain , Disease Models, Animal , Dose-Response Relationship, Drug , Fascia/physiopathology , Male , Nerve Growth Factor/pharmacology , Nociceptive Pain/chemically induced , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
7.
Toxicol Appl Pharmacol ; 414: 115428, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33524449

ABSTRACT

Researches have shown that calcitonin gene-related peptide (CGRP) plays a pivotal role in pain modulation. Nociceptive information from the periphery is relayed from parabrachial nucleus (PBN) to brain regions implicated involved in pain. This study investigated the effects and mechanisms of CGRP and CGRP receptors in pain regulation in the PBN of naive and neuropathic pain rats. Chronic sciatic nerve ligation was used to model neuropathic pain, CGRP and CGRP 8-37 were injected into the PBN of the rats, and calcitonin receptor-like receptor (CLR), a main structure of CGRP receptor, was knocked down by lentivirus-coated CLR siRNA. The hot plate test (HPT) and the Randall Selitto Test (RST) was used to determine the latency of the rat hindpaw response. The expression of CLR was detected with RT-PCR and western blotting. We found that intra-PBN injecting of CGRP induced an obvious anti-nociceptive effect in naive and neuropathic pain rats in a dose-dependent manner, the CGRP-induced antinociception was significantly reduced after injection of CGRP 8-37, Moreover, the mRNA and protein levels of CLR, in PBN decreased significantly and the antinociception CGRP-induced was also significantly lower in neuropathic pain rats than that in naive rats. Knockdown CLR in PBN decreased the expression of CLR and the antinociception induced by CGRP was observably decreased. Our results demonstrate that CGRP induced antinociception in PBN of naive or neuropathic pain rats, CGRP receptor mediates this effect. Neuropathic pain induced decreases in the expression of CGRP receptor, as well as in CGRP-induced antinociception in PBN.


Subject(s)
Analgesics/pharmacology , Calcitonin Gene-Related Peptide/pharmacology , Calcitonin Receptor-Like Protein/agonists , Nociceptive Pain/prevention & control , Pain Threshold/drug effects , Parabrachial Nucleus/drug effects , Peptide Fragments/pharmacology , Receptors, Calcitonin Gene-Related Peptide/agonists , Sciatica/prevention & control , Animals , Calcitonin Receptor-Like Protein/genetics , Calcitonin Receptor-Like Protein/metabolism , Disease Models, Animal , Gene Expression Regulation , Male , Nociceptive Pain/genetics , Nociceptive Pain/metabolism , Nociceptive Pain/physiopathology , Parabrachial Nucleus/metabolism , Parabrachial Nucleus/physiopathology , Rats, Sprague-Dawley , Receptors, Calcitonin Gene-Related Peptide/genetics , Receptors, Calcitonin Gene-Related Peptide/metabolism , Sciatica/genetics , Sciatica/metabolism , Sciatica/physiopathology
8.
Toxicol Appl Pharmacol ; 416: 115458, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33607128

ABSTRACT

Morphine is an opioid agonist and a nonselective mu, kappa and delta receptor agonist. It is a commonly used analgesic drug for the treatment of acute and chronic pain as well as cancer pain. Morphine is particularly important to address the problem of morphine tolerance. Tcf7l2, known as a risk gene for schizophrenia and autism, encodes a member of the LEF1/TCF transcription factor family. TCF7L2 is an important transcription factor that is upregulated in neuropathic pain models. However, the relationship between TCF7L2 and morphine tolerance has not been reported. In this study, we found that morphine tolerance led to the upregulation of TCF7L2 in the spinal cord, and also led to the upregulation of TCF7L2 expression in glial cells, which promoted inflammation related signal, and activated TLR4 / NF-κB/NLRP3 pathway. In addition, TCF7L2 regulated microglial cell activation induced by chronic morphine treatment. Mechanically, we found that TCF7L2 transcriptionally regulated TLR4 expression, and the depletion of TCF7L2 alleviated morphine tolerance induced by chronic morphine treatment, and further alleviated pain hypersensitivity induced by chronic morphine treatment. We therefore suggested that TCF7L2 regulates the activation of TLR4/ NF-κB/NLRP3 pathway in microglia, and is involved in the formation of morphine tolerance. Our results provide a new idea for the regulation mechanism of morphine tolerance.


Subject(s)
Analgesics, Opioid/toxicity , Drug Tolerance , Hyperalgesia/chemically induced , Microglia/drug effects , Morphine/toxicity , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nociceptive Pain/prevention & control , Toll-Like Receptor 4/metabolism , Transcription Factor 7-Like 2 Protein/metabolism , Animals , Cell Line , Disease Models, Animal , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Male , Mice , Microglia/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Nociceptive Pain/metabolism , Nociceptive Pain/physiopathology , Pain Threshold/drug effects , Signal Transduction , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/physiopathology , Toll-Like Receptor 4/genetics , Transcription Factor 7-Like 2 Protein/genetics , Up-Regulation
9.
Anesth Analg ; 132(2): 406-419, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33332902

ABSTRACT

A serious adverse effect of prescription opioid analgesics is addiction, both to these analgesics and to illicit drugs like heroin that also activate the µ-opioid receptor (MOR). Opioid use disorder (OUD) and opioid overdose deaths represent a current American health crisis, and the prescription of opioid analgesics has contributed significantly to this crisis. While prescription opioids are highly effective analgesics, there currently exists no facile way to use them for extended periods without the risk of addiction. If addiction caused by MOR-targeting analgesics could be blocked by blending in a new "antiaddiction" ingredient that does not diminish analgesia and does not introduce its own therapeutically limiting side effects, then continued clinical use of prescription opioids for treating pain could be maintained (or even enhanced) instead of curtailed. In this narrative review, we contextualize this hypothesis, first with a brief overview of the current American opioid addiction crisis. The neurobiology of 2 key receptors in OUD development, MOR and the κ-opioid receptor (KOR), is then discussed to highlight the neuroanatomical features and circuitry in which signal transduction from these receptors lie in opposition-creating opportunities for pharmacological intervention in curtailing the addictive potential of MOR agonism. Prior findings with mixed MOR/KOR agonists are considered before exploring new potential avenues such as biased KOR agonists. New preclinical data are highlighted, demonstrating that the G protein-biased KOR agonist nalfurafine reduces the rewarding properties of MOR-targeting analgesics and enhances MOR-targeting analgesic-induced antinociception. Finally, we discuss the recent discovery that a regulator of G protein signaling (namely, RGS12) is a key component of signaling bias at KOR, presenting another drug discovery target toward identifying a single agent or adjuvant to be added to traditional opioid analgesics that could reduce or eliminate the addictive potential of the latter drug.


Subject(s)
Drug Design , Narcotic Antagonists/pharmacology , Nociception/drug effects , Nociceptive Pain/drug therapy , Opioid-Related Disorders/prevention & control , Receptors, Opioid, kappa/agonists , Receptors, Opioid, mu/agonists , Animals , Humans , Molecular Structure , Narcotic Antagonists/adverse effects , Narcotic Antagonists/chemistry , Nociceptive Pain/metabolism , Nociceptive Pain/physiopathology , Nociceptive Pain/psychology , Opioid-Related Disorders/etiology , RGS Proteins/metabolism , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Signal Transduction , Structure-Activity Relationship
10.
Int J Mol Sci ; 22(2)2021 Jan 06.
Article in English | MEDLINE | ID: mdl-33418928

ABSTRACT

Free nerve endings are key structures in sensory transduction of noxious stimuli. In spite of this, little is known about their functional organization. Transient receptor potential (TRP) channels have emerged as key molecular identities in the sensory transduction of pain-producing stimuli, yet the vast majority of our knowledge about sensory TRP channel function is limited to data obtained from in vitro models which do not necessarily reflect physiological conditions. In recent years, the development of novel optical methods such as genetically encoded calcium indicators and photo-modulation of ion channel activity by pharmacological tools has provided an invaluable opportunity to directly assess nociceptive TRP channel function at the nerve terminal.


Subject(s)
Nociceptive Pain/pathology , Peripheral Nerves/metabolism , Transient Receptor Potential Channels/metabolism , Animals , Axons/metabolism , Calcium Signaling/drug effects , Capsaicin/pharmacology , Nociceptive Pain/metabolism , Precision Medicine , Sensory Receptor Cells/metabolism , Transient Receptor Potential Channels/antagonists & inhibitors
11.
J Pharmacol Exp Ther ; 375(1): 104-114, 2020 10.
Article in English | MEDLINE | ID: mdl-32759368

ABSTRACT

This dose-response study investigated the effects of sialorphin on [Met5]enkephalin (ME)-induced inhibition of contractions in mouse vas deferens and antinociception in male rats. Differences were compared among combinations of three chemical peptidase inhibitors: amastatin, captopril, and phosphoramidon. The ratio of potencies of ME in mouse vas deferens pretreated with both sialorphin (100 µM) and a mixture of the three peptidase inhibitors (1 µM each) was higher than that with the mixture of peptidase inhibitors alone at any dose. Intrathecal administration of sialorphin (100-400 nmol) significantly and dose dependently increased ME (3 nmol)-induced antinociception with the mixture of three peptidase inhibitors (10 nmol each). The degree of antinociception with a combination of any two of the peptidase inhibitors (10 nmol each) in the absence of sialorphin was less than that in the presence of sialorphin (200 nmol). Pretreatment with both sialorphin (200 nmol) and the mixture of three peptidase inhibitors (10 nmol each) produced an approximately 100-fold augmentation in ME (10 nmol)-induced antinociception, but without signs of toxicity such as motor dysfunction in rats. Radioligand receptor binding assay revealed that sialorphin did not affect either binding affinity or maximal binding capacity of [d-Ala2,N-MePhe4,Gly-ol5]enkephalin. These results indicate that sialorphin potentiates the effects of ME without toxicity by a mechanism other than peptidase inhibition and with no effect on its affinity to µ-opioid receptors. SIGNIFICANCE STATEMENT: Sialorphin is regarded as an endogenous peptidase inhibitor that interacts with enkephalin-degrading enzymes. The results of these in vitro and in vivo studies confirm that sialorphin potentiates the effects of [Met5]enkephalin without toxicity by an action other than peptidase inhibition. This suggests that sialorphin offers the advantage of reducing or negating the side effects of opioid drugs and endogenous opioid peptides.


Subject(s)
Analgesics/pharmacology , Enkephalin, Methionine/pharmacology , Peptides/pharmacology , Protease Inhibitors/pharmacology , Vas Deferens/drug effects , Analgesics/administration & dosage , Animals , Dose-Response Relationship, Drug , Drug Synergism , Enkephalin, Methionine/administration & dosage , In Vitro Techniques , Injections, Spinal , Male , Mice , Mice, Inbred ICR , Motor Activity/drug effects , Nociceptive Pain/drug therapy , Nociceptive Pain/metabolism , Pain Measurement , Peptides/administration & dosage , Protease Inhibitors/administration & dosage , Protein Binding , Radioligand Assay , Rats, Wistar , Receptors, Opioid/metabolism
12.
Anesthesiology ; 133(6): 1244-1259, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32997750

ABSTRACT

BACKGROUND: Acid-sensing ion channel 3 (ASIC3) upregulation has been reported in dorsal root ganglion neurons after incision and contributes to postoperative nociception. This study hypothesized that upregulation of ASIC3 in incised tissues is induced by nerve growth factor through the phosphoinositide 3-kinase/protein kinase B signaling pathway. METHODS: A plantar incision model was established in adult male and female Sprague-Dawley rats. ASIC3 was inhibited by APETx2 treatment, small interfering RNA treatment, or ASIC3 knockout. Sciatic nerve ligation was performed to analyze ASIC3 transport. A nerve growth factor antibody and a phosphoinositide 3-kinase inhibitor were used to investigate the mechanism by which nerve growth factor regulates ASIC3 expression. RESULTS: Acid-sensing ion channel 3 inhibition decreased incisional guarding and mechanical nociception. ASIC3 protein levels were increased in skin and muscle 4 h after incision (mean ± SD: 5.4 ± 3.2-fold in skin, n = 6, P = 0.001; 4.3 ± 2.2-fold in muscle, n = 6, P = 0.001). Sciatic nerve ligation revealed bidirectional ASIC3 transport. Nerve growth factor antibody treatment inhibited the expression of ASIC3 (mean ± SD: antibody 2.3 ± 0.8-fold vs. vehicle 4.9 ± 2.4-fold, n = 6, P = 0.036) and phosphorylated protein kinase B (mean ± SD: antibody 0.8 ± 0.3-fold vs. vehicle 1.8 ± 0.8-fold, n = 6, P = 0.010) in incised tissues. Intraplantar injection of nerve growth factor increased the expression of ASIC3 and phosphorylated protein kinase B. ASIC3 expression and incisional pain-related behaviors were inhibited by pretreatment with the phosphoinositide 3-kinase inhibitor LY294002. CONCLUSIONS: Acid-sensing ion channel 3 overexpression in incisions contributes to postoperative guarding and mechanical nociception. Bidirectional transport of ASIC3 between incised tissues and dorsal root ganglion neurons occurs through the sciatic nerve. Nerve growth factor regulates ASIC3 expression after plantar incision through the phosphoinositide 3-kinase/protein kinase B signaling pathway.


Subject(s)
Acid Sensing Ion Channels/metabolism , Nerve Growth Factor/metabolism , Nociceptive Pain/metabolism , Nociceptive Pain/physiopathology , Pain, Postoperative/metabolism , Pain, Postoperative/physiopathology , Animals , Disease Models, Animal , Female , Male , Rats , Rats, Sprague-Dawley , Signal Transduction
13.
Cell Mol Life Sci ; 76(10): 1889-1899, 2019 May.
Article in English | MEDLINE | ID: mdl-30788514

ABSTRACT

Chronic neuropathic pain is a debilitating condition that remains challenging to treat. Glutamate N-methyl-D-aspartate receptor (NMDAR) antagonists have been used to treat neuropathic pain, but the exact sites of their actions have been unclear until recently. Although conventionally postsynaptic, NMDARs are also expressed presynaptically, particularly at the central terminals of primary sensory neurons, in the spinal dorsal horn. However, presynaptic NMDARs in the spinal cord are normally quiescent and are not actively involved in physiological nociceptive transmission. In this review, we describe the emerging role of presynaptic NMDARs at the spinal cord level in chronic neuropathic pain and the implications of molecular mechanisms for more effective treatment. Recent studies indicate that presynaptic NMDAR activity at the spinal cord level is increased in several neuropathic pain conditions but not in chronic inflammatory pain. Increased presynaptic NMDAR activity can potentiate glutamate release from primary afferent terminals to spinal dorsal horn neurons, which is crucial for the synaptic plasticity associated with neuropathic pain caused by traumatic nerve injury and chemotherapy-induced peripheral neuropathy. Furthermore, α2δ-1, previously considered a calcium channel subunit, can directly interact with NMDARs through its C-terminus to increase presynaptic NMDAR activity by facilitating synaptic trafficking of α2δ-1-NMDAR complexes in neuropathic pain caused by chemotherapeutic agents and peripheral nerve injury. Targeting α2δ-1-bound NMDARs with gabapentinoids or α2δ-1 C-terminus peptides can attenuate nociceptive drive form primary sensory nerves to dorsal horn neurons in neuropathic pain.


Subject(s)
Neuralgia/physiopathology , Nociceptive Pain/physiopathology , Receptors, N-Methyl-D-Aspartate/physiology , Receptors, Presynaptic/physiology , Spinal Cord/physiopathology , Animals , Mice , Neuralgia/metabolism , Nociception/physiology , Nociceptive Pain/metabolism , Nociceptors/metabolism , Nociceptors/physiology , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Presynaptic/metabolism
14.
Neural Plast ; 2020: 8885729, 2020.
Article in English | MEDLINE | ID: mdl-33144854

ABSTRACT

Depression and pain disorders share a high degree of comorbidity. Inflammatory mechanisms play an important role in the pathogenesis of depression-chronic somatic pain comorbidity. In this study, we investigated the effects of acupuncture on blood and brain regional tumor necrosis factor alpha (TNF-α) in rats with depression and chronic somatic pain comorbidity. Forty Sprague-Dawley rats were randomly divided into the following 4 groups with 10 each: control, model, model treated with transcutaneous auricular vagus nerve stimulation (taVNS), and model treated with electroacupuncture (EA). Chronic unpredictable mild stress (CUMS) with chronic constriction injury of the sciatic nerve (CCI) was used to produce depression and chronic somatic pain comorbidity in the latter 3 groups. The rats of the taVNS and EA groups received, respectively, taVNS and EA at ST 36 for 28 days. Pain intensity was measured using a mechanical withdrawal threshold and thermal stimulation latency once biweekly. Depressive behavior was examined using a sucrose preference test at baseline and the end of modeling and intervention. The level of plasma TNF-α and the expression of TNF-α in the prefrontal cortex (PFC), hippocampus, amygdala, and hypothalamus were measured. While CUMS plus CCI produced remarkable depression-like behavior and pain disorders, EA and taVNS significantly improved depression and reduced pain intensity. CUMS plus CCI also resulted in a significant increase in plasma TNF-α level and the expression in all brain regions examined compared to the intact controls. Both EA and taVNS interventions, however, suppressed the elevated level of TNF-α. These results suggest that EA and taVNS have antidepressant and analgesic effects. Such effects may be associated with the suppression of TNF-α-related neuroinflammation.


Subject(s)
Brain/metabolism , Depression/metabolism , Nociceptive Pain/metabolism , Transcutaneous Electric Nerve Stimulation , Tumor Necrosis Factor-alpha/metabolism , Vagus Nerve Stimulation , Acupuncture Therapy , Animals , Male , Rats, Sprague-Dawley
15.
Int J Mol Sci ; 21(24)2020 Dec 18.
Article in English | MEDLINE | ID: mdl-33353118

ABSTRACT

Despite the availability of the current drug arsenal for pain management, there is still a clinical need to identify new, more effective, and safer analgesics. Based on our earlier study, newly synthesized 1,3,4-oxadiazole derivatives of pyrrolo[3,4-d]pyridazinone, especially 10b and 13b, seem to be promising as potential analgesics. The current study was designed to investigate whether novel derivatives attenuate nociceptive response in animals subjected to thermal or chemical noxious stimulus, and to compare this effect to reference drugs. The antinociceptive effect of novel compounds was studied using the tail-flick and formalin test. Pretreatment with novel compounds at all studied doses increased the latency time in the tail-flick test and decreased the licking time during the early phase of the formalin test. New derivatives given at the medium and high doses also reduced the late phase of the formalin test. The achieved results indicate that new derivatives dose-dependently attenuate nociceptive response in both models of pain and exert a lack of gastrotoxicity. Both studied compounds act more efficiently than indomethacin, but not morphine. Compound 13b at the high dose exerts the greatest antinociceptive effect. It may be due to the reduction of nociceptor sensitization via prostaglandin E2 and myeloperoxidase levels decrease.


Subject(s)
Analgesics/pharmacology , Gastric Mucosa/drug effects , Nociception/drug effects , Nociceptive Pain/drug therapy , Oxadiazoles/chemistry , Pyridazines/chemistry , Pyrroles/chemistry , Analgesics/chemistry , Animals , Dinoprostone/metabolism , Gastric Mucosa/pathology , Male , Nociceptive Pain/metabolism , Nociceptive Pain/pathology , Pain Measurement , Peroxidase/metabolism , Rats , Rats, Wistar
16.
Am J Physiol Renal Physiol ; 317(1): F90-F98, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31091120

ABSTRACT

Altered Toll-like receptor (TLR)4 activation has been identified in several chronic pain conditions but has not been well studied in interstitial cystitis/bladder pain syndrome (IC/BPS). Our previously published human studies indicated that patients with IC/BPS present altered systemic TLR4-mediated inflammatory responses, which were significantly correlated with reported pain severity. In the present study, we sought to determine whether altered TLR4 activation plays a role in pelvic/bladder pain seen in patients with IC/BPS using our validated IC/BPS-like transgenic autoimmune cystitis model (URO-OVA). URO-OVA mice developed responses consistent with pelvic and bladder pain after cystitis induction, which was associated with increased splenocyte production of TLR4-mediated proinflammatory cytokines IL-1ß, IL-6, and TNF-α. Increased spinal expression of mRNAs for proinflammatory cytokines IL-6 and TNF-α, glial activation markers CD11b and glial fibrillary acidic protein, and endogenous TLR4 ligand high mobility group box 1 was also observed after cystitis induction. Compared with URO-OVA mice, TLR4-deficient URO-OVA mice developed significantly reduced nociceptive responses, although similar bladder inflammation and voiding dysfunction, after cystitis induction. Intravenous administration of TAK-242 (a TLR4-selective antagonist) significantly attenuated nociceptive responses in cystitis-induced URO-OVA mice, which was associated with reduced splenocyte production of TLR4-mediated IL-1ß, IL-6, and TNF-α as well as reduced spinal expression of mRNAs for IL-6, TNF-α, CD11b, glial fibrillary acidic protein, and high mobility group box 1. Our results indicate that altered TLR4 activation plays a critical role in bladder nociception independent of inflammation and voiding dysfunction in the URO-OVA model, providing a potential mechanistic insight and therapeutic target for IC/BPS pain.


Subject(s)
Autoimmune Diseases/metabolism , Cystitis, Interstitial/metabolism , Nociceptive Pain/metabolism , Pain Threshold , Toll-Like Receptor 4/metabolism , Urinary Bladder/metabolism , Analgesics/pharmacology , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/physiopathology , Cells, Cultured , Cystitis, Interstitial/genetics , Cystitis, Interstitial/immunology , Cystitis, Interstitial/physiopathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Inflammation Mediators/metabolism , Mice, Inbred C57BL , Mice, Knockout , Nociceptive Pain/genetics , Nociceptive Pain/immunology , Nociceptive Pain/physiopathology , Ovalbumin/genetics , Ovalbumin/immunology , Ovalbumin/metabolism , Pain Threshold/drug effects , Signal Transduction , Spine/immunology , Spine/metabolism , Spleen/immunology , Spleen/metabolism , Sulfonamides/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Urinary Bladder/drug effects , Urinary Bladder/immunology , Urinary Bladder/physiopathology , Urodynamics
17.
Toxicol Appl Pharmacol ; 369: 30-38, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30763598

ABSTRACT

Despite all the development of modern medicine, around 100 compounds derived from natural products were undergoing clinical trials only at the end of 2013. Among these natural substances in clinical trials, we found the resveratrol (RES), a pharmacological multi-target drug. RES analgesic properties have been demonstrated, although the bases of these mechanisms have not been fully elucidated. The aim of this study was to evaluate the involvement of opioid and cannabinoid systems in RES-induced peripheral antinociception. Paw withdrawal method was used and hyperalgesia was induced by carrageenan (200 µg/paw). All drugs were given by intraplantar injection in male Swiss mice (n = 5). RES (100 µg/paw) administered in the right hind paw induced local antinociception that was antagonized by naloxone, non-selective opioid receptor antagonist, and clocinnamox, µOR selective antagonist. Naltrindole and nor-binaltorfimine, selective antagonists for δOR and kOR, respectively, did not reverse RES-induced peripheral antinociception. CB1R antagonist AM251, but not CB2R antagonist AM630, antagonized RES-induced peripheral antinociception. Peripheral antinociception of RES intermediate-dose (50 µg/paw) was increased by: (i) bestatin, inhibitor of endogenous opioid degradation involved-enzymes; (ii) MAFP, inhibitor of anandamide amidase; (iii) JZL184, inhibitor of 2-arachidonoylglycerol degradation involved-enzyme; (iv) VDM11, endocannabinoid reuptake inhibitor. Acute and peripheral administration of RES failed to affect the amount of µOR, CB1R and CB2R. Experimental data suggest that RES induces peripheral antinociception through µOR and CB1R activation by endogenous opioid and endocannabinoid releasing.


Subject(s)
Analgesics/pharmacology , Endocannabinoids/metabolism , Hyperalgesia/prevention & control , Nociceptive Pain/prevention & control , Opioid Peptides/metabolism , Receptor, Cannabinoid, CB1/agonists , Receptors, Opioid, mu/agonists , Resveratrol/pharmacology , Animals , Behavior, Animal/drug effects , Cannabinoid Receptor Antagonists/pharmacology , Carrageenan , Disease Models, Animal , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Hyperalgesia/psychology , Male , Mice , Narcotic Antagonists/pharmacology , Nociceptive Pain/chemically induced , Nociceptive Pain/metabolism , Nociceptive Pain/psychology , Receptor, Cannabinoid, CB1/metabolism , Receptors, Opioid, mu/metabolism , Signal Transduction
18.
Mol Psychiatry ; 23(8): 1745-1755, 2018 08.
Article in English | MEDLINE | ID: mdl-28485408

ABSTRACT

Development of an efficacious, non-addicting analgesic has been challenging. Discovery of novel mechanisms underlying addiction may present a solution. Here we target the neurokinin system, which is involved in both pain and addiction. Morphine exerts its rewarding actions, at least in part, by inhibiting GABAergic input onto substance P (SP) neurons in the ventral tegmental area (VTA), subsequently increasing SP release onto dopaminergic neurons. Genome editing of the neurokinin 1 receptor (NK1R) in the VTA renders morphine non-rewarding. Complementing our genetic approach, we demonstrate utility of a bivalent pharmacophore with dual activity as a µ/δ opioid agonist and NK1R antagonist in inhibiting nociception in an animal model of acute pain while lacking any positive reinforcement. These data indicate that dual targeting of the dopaminergic reward circuitry and pain pathways with a multifunctional opioid agonist-NK1R antagonist may be an efficacious strategy in developing future analgesics that lack abuse potential.


Subject(s)
Neurokinin-1 Receptor Antagonists/pharmacology , Opioid-Related Disorders/prevention & control , Receptors, Neurokinin-1/metabolism , Acute Pain/drug therapy , Acute Pain/metabolism , Analgesics/pharmacology , Animals , CRISPR-Cas Systems , Disease Models, Animal , Dopamine/metabolism , Escherichia coli , Gene Knockdown Techniques , Male , Mice, Inbred ICR , Morphine/pharmacology , Nociceptive Pain/drug therapy , Nociceptive Pain/metabolism , Opioid-Related Disorders/genetics , Opioid-Related Disorders/metabolism , Rats, Sprague-Dawley , Receptors, Neurokinin-1/genetics , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Reward , Substance P/metabolism , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
19.
Behav Pharmacol ; 30(6): 534-537, 2019 09.
Article in English | MEDLINE | ID: mdl-31033524

ABSTRACT

Chronic neuropathic pain is a burden to millions of patients every day. Patients with neuropathic pain will also experience acute pain throughout their everyday lives adding to their nociceptive burden. Using nociceptive models in mice this study aimed to investigate the relationship between acute visceral pain and chronic neuropathic pain in spontaneous and affective behaviors. Neuropathic pain was induced by chronic constriction injury (CCI) of the sciatic nerve of C57BL/6J male mice and examined in assays of acetic acid (AA)-induced stretching or conditioned place aversion to assess nociceptive and aversive behaviors. Stretching induced by a low concentration (0.32%) of AA given intraperitoneally was significantly increased in CCI and paclitaxel-treated animals compared to control animals. A higher concentration (1.2%) of AA was able to induce stretching equally in both neuropathic and control mice. In the conditioned place aversion test, an AA concentration of 0.32% did not induce place aversion in either sham or CCI animals. However, the 1.2% concentration of AA-induced higher place aversion scores in CCI mice compared to sham mice. No difference in place conditioning was observed between paclitaxel and vehicle-treated mice. Overall, our results show that peripheral nerve injury and paclitaxel treatment induces hypersensitivity to AA-induced nociception and place aversion.


Subject(s)
Neuralgia/physiopathology , Neuralgia/psychology , Nociceptive Pain/physiopathology , Acetic Acid/metabolism , Acetic Acid/pharmacology , Animals , Conditioning, Classical , Disease Models, Animal , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Male , Mice , Mice, Inbred C57BL , Nociception/physiology , Nociceptive Pain/metabolism , Sciatic Nerve/injuries
20.
J Pharmacol Sci ; 140(3): 310-312, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31492577

ABSTRACT

We tested whether genetic deletion of Cav3.2 T-type Ca2+ channels abolishes hydrogen sulfide (H2S)-mediated pain signals in mice. In Cav3.2-expressing HEK293 cells, Na2S, an H2S donor, at 100 µM clearly increased Ba2+ currents, as assessed by whole-cell patch-clamp recordings. In wild-type C57BL/6 mice, intraplantar and intracolonic administration of Na2S evoked mechanical allodynia and visceral nociceptive behavior, respectively, which were abolished by TTA-A2, a T-type Ca2+ channel blocker. In Cav3.2-knockout mice of a C57BL/6 background, Na2S caused neither somatic allodynia nor colonic nociception. Our study thus provides definitive evidence for an essential role of Cav3.2 in H2S-dependent somatic and colonic pain.


Subject(s)
Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/metabolism , Hydrogen Sulfide/pharmacology , Nociceptive Pain/metabolism , Signal Transduction/drug effects , Visceral Pain/metabolism , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cell Line , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar , TRPA1 Cation Channel/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL