ABSTRACT
Neuropathic pain is a condition with varying origins, including reduced dietary micronutrient intake. Phytate is a polyphosphate found in seeds and grains that can act as an antinutrient due to the ability of sequester essential divalent metals. Here we tested whether moderate dietary phytate intake could alter nociceptive pain. We subjected weaning mice to a chow supplemented with 1% phytate for eight weeks. Body weight gain, glycemic responses, food ingestion, water ingestion, and liver and adipose tissue weights were not altered compared to controls. We observed a decreased mechanical allodynia threshold in the intervention group, although there were no changes in heat- or cold-induced pain. Animals consuming phytate showed reduced spinal cord tumor necrosis factor (TNF), indicating altered inflammatory process. These data provide evidence for a subclinical induction of mechanical allodynia that is independent of phytate consumption in animals with otherwise normal phenotypic pattern.
Subject(s)
Hyperalgesia , Neuralgia , Mice , Animals , Hyperalgesia/etiology , Phytic Acid , Spinal Cord , Tumor Necrosis Factor-alphaABSTRACT
BACKGROUND: Fibromyalgia overlaps and/or mimics other rheumatic diseases and may be a confounding factor in the clinimetric assessment of these illnesses. Allodynia is a distinctive fibromyalgia feature that can be elicited during routine blood pressure measurement. For epidemiological purposes fibromyalgia can be diagnosed using the 2016 Wolfe et al. criteria questionnaire. No physical examination is required. OBJECTIVE: To evaluate the role of a straightforward question formulated during routine blood pressure measurement for fibromyalgia detection in a rheumatology outpatient clinic. PATIENTS AND METHODS: All adult patients attending our Rheumatology outpatient clinic were invited to participate. While awaiting their medical consultation, they filled-out the 2016 Wolfe et al. FM diagnostic criteria questionnaire. During the ensuing routine physical examination, the physician advanced the following guideline: "I am going to take your blood pressure; tell me if the cuff's pressure causes pain". Then, blood pressure cuff was inflated to 170 mm/Hg. Sphygmomanometry induced allodynia was defined as any local discomfort caused by blood pressure measurement. If a patient voiced any uneasiness, a follow-up dichotomic question was formulated "did it hurt much or little". Sphygmomanometry-induced allodynia was correlated with the presence of fibromyalgia according to the 2016 Wolfe diagnostic criteria. RESULTS: Four hundred and ninety-one patients were included in the study; most of them (84%) were female. The female cohort displayed the following features: Twenty five percent had fibromyalgia. Twenty seven percent had sphygmomanometry-induced allodynia. In women, sphygmomanometry-evoked allodynia had 63% sensitivity and 84% specificity for fibromyalgia diagnosis. The area under curve was 0.751. Moreover, having "much" local pain elicitation during blood pressure testing had 23% sensitivity and 96% specificity for fibromyalgia diagnosis. Men behaved differently; 15% fulfilled the fibromyalgia diagnostic criteria, but only 2% had sphygmomanometry induced allodynia. CONCLUSIONS: Inquiring female patients about local discomfort during routine blood pressure measurement is a simple and efficient procedure for fibromyalgia detection. This undemanding approach could be implemented in all clinical settings. There is marked sexual dimorphism in the link between sphygmomanometry-induced allodynia and fibromyalgia diagnosis. The presence of fibromyalgia is almost certain in those individuals having substantial pain elicitation during blood pressure measurement.
Subject(s)
Fibromyalgia , Adult , Male , Humans , Female , Fibromyalgia/complications , Fibromyalgia/diagnosis , Cross-Sectional Studies , Hyperalgesia/diagnosis , Hyperalgesia/etiology , Blood Pressure , Pain Measurement/methods , Pain , Surveys and QuestionnairesABSTRACT
Chronic stress affects millions of people around the world, and it can trigger different behavioral disorders like nociceptive hypersensitivity and anxiety, among others. However, the mechanisms underlaying these chronic stress-induced behavioral disorders have not been yet elucidated. This study was designed to understand the role of high-mobility group box-1 (HMGB1) and toll-like receptor 4 (TLR4) in chronic stress-induced nociceptive hypersensitivity. Chronic restraint stress induced bilateral tactile allodynia, anxiety-like behaviors, phosphorylation of extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase (p38MAPK) and activation of spinal microglia. Moreover, chronic stress enhanced HMGB1 and TLR4 protein expression at the dorsal root ganglion, but not at the spinal cord. Intrathecal injection of HMGB1 or TLR4 antagonists reduced tactile allodynia and anxiety-like behaviors induced by chronic stress. Additionally, deletion of TLR4 diminished the establishment of chronic stress-induced tactile allodynia in male and female mice. Lastly, the antiallodynic effect of HMGB1 and TLR4 antagonists were similar in stressed male and female rats and mice. Our results suggest that chronic restraint stress induces nociceptive hypersensitivity, anxiety-like behaviors, and up-regulation of spinal HMGB1 and TLR4 expression. Blockade of HMGB1 and TLR4 reverses chronic restraint stress-induced nociceptive hypersensitivity and anxiety-like behaviors and restores altered HMGB1 and TLR4 expression. The antiallodynic effects of HMGB1 and TLR4 blockers in this model are sex independent. TLR4 could be a potential pharmacological target for the treatment of the nociceptive hypersensitivity associated with widespread chronic pain.
Subject(s)
HMGB1 Protein , Hyperalgesia , Animals , Female , Male , Mice , Rats , Alarmins/metabolism , Chronic Disease , HMGB1 Protein/metabolism , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Hyperalgesia/metabolism , Nociception , p38 Mitogen-Activated Protein Kinases/metabolism , Spinal Cord , Toll-Like Receptor 4/metabolismABSTRACT
Resolvin D5 (RvD5) is a specialized pro-resolving lipid mediator with potent anti-inflammatory and analgesic properties. Orofacial pain conditions, especially those that are chronic, present clinical challenges in terms of pharmacological management. Thus, new therapeutic options are clearly warranted. Herein, we investigated the antinociceptive effect of RvD5 in the chronic constriction injury of the infraorbital nerve (CCI-ION) model and in the orofacial formalin test in female and male Wistar rats. Our results indicated that repeated subarachnoid medullary injections of RvD5 at 10 ng resulted in a significant reduction of heat and mechanical hyperalgesia induced by the CCI-ION in male and female rats, but males were more sensitive to RvD5 effects. In addition, after CCI-ION, interleukin-6 (IL-6) level was increased in the trigeminal nucleus caudalis of male, but not female rats, which was reduced by RvD5 repeated treatment. No changes in the levels of IL-1ß were found. Minocycline blocked the effect of RvD5 in male rats but failed to affect RvD5 antinociceptive effect in females. Moreover, a single medullary injection of RvD5 caused a significant reduction of formalin-induced facial grooming, in phases I and II of the test, but only in male rats. This study demonstrated for the first time the analgesic effect of RvD5 in trigeminal pain models, and corroborated previous evidence of sex dichotomy, with a greater effect in males. This article presents a translational potential of RvD5 for targeted therapies aiming at the control of acute and chronic trigeminal pain, but further studies are needed to elucidate its sex-related mechanisms. PERSPECTIVE: This study demonstrated that RvD5 may provide the benefits for trigeminal neuropathic pain treatment in male and female rats, but its effect on inflammatory orofacial pain seems to be restricted only to males. Also, it provided the evidence for sex dichotomy in the mechanisms related to the antinociceptive effect of RvD5.
Subject(s)
Analgesia , Chronic Pain , Female , Rats , Male , Animals , Sex Characteristics , Rats, Sprague-Dawley , Rats, Wistar , Facial Pain/drug therapy , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Analgesics/therapeutic use , Chronic Pain/drug therapy , Disease Models, AnimalABSTRACT
Pannexin 1 (Panx1) is involved in the spinal central sensitization process in rats with neuropathic pain, but its interaction with well-known, pain-related, ligand-dependent receptors, such as NMDA receptors (NMDAR) and P2X7 purinoceptors (P2X7R), remains largely unexplored. Here, we studied whether NMDAR- and P2X7R-dependent nociceptive signaling in neuropathic rats require the activation of Panx1 channels to generate spinal central sensitization, as assessed by behavioral (mechanical hyperalgesia) and electrophysiological (C-reflex wind-up potentiation) indexes. Administration of either a selective NMDAR agonist i.t. (NMDA, 2 mM) or a P2X7R agonist (BzATP, 150 µM) significantly increased both the mechanical hyperalgesia and the C-reflex wind-up potentiation, effects that were rapidly reversed (minutes) by i.t. administration of a selective pannexin 1 antagonist (10panx peptide, 300 µM), with the scores even reaching values of rats without neuropathy. Accordingly, 300 µM 10panx completely prevented the effects of NMDA and BzATP administered 1 h later, on mechanical hyperalgesia and C-reflex wind-up potentiation. Confocal immunofluorescence imaging revealed coexpression of Panx1 with NeuN protein in intrinsic dorsal horn neurons of neuropathic rats. The results indicate that both NMDAR- and P2X7R-mediated increases in mechanical hyperalgesia and C-reflex wind-up potentiation require neuronal Panx1 channel activation to initiate and maintain nociceptive signaling in neuropathic rats.
Subject(s)
Connexins/metabolism , Hyperalgesia , Nerve Tissue Proteins/metabolism , Receptors, Purinergic P2X7 , Animals , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Hyperalgesia/metabolism , N-Methylaspartate/metabolism , Nociception , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Purinergic P2X7/metabolism , Spinal Cord/metabolismABSTRACT
ABSTRACT: Diabetic neuropathy, often associated with diabetes mellitus, is a painful condition with no known effective treatment except glycemic control. Studies with neuropathic pain models report alterations in cannabinoid and opioid receptor expression levels; receptors whose activation induces analgesia. We examined whether interactions between CB1R and opioid receptors could be targeted for the treatment of diabetic neuropathy. For this, we generated antibodies that selectively recognize native CB1R-MOR and CB1R-DOR heteromers using a subtractive immunization strategy. We assessed the levels of CB1R, MOR, DOR, and interacting complexes using a model of streptozotocin-induced diabetic neuropathy and detected increased levels of CB1R, MOR, DOR, and CB1R-MOR complexes compared with those in controls. An examination of G-protein signaling revealed that activity induced by the MOR, but not the DOR agonist, was potentiated by low nanomolar doses of CB1R ligands, including antagonists, suggesting an allosteric modulation of MOR signaling by CB1R ligands within CB1R-MOR complexes. Because the peptide endocannabinoid, hemopressin, caused a significant potentiation of MOR activity, we examined its effect on mechanical allodynia and found that it blocked allodynia in wild-type mice and mice with diabetic neuropathy lacking DOR (but have CB1R-MOR complexes). However, hemopressin does not alter the levels of CB1R-MOR complexes in diabetic mice lacking DOR but increases the levels of CB1R-DOR complexes in diabetic mice lacking MOR. Together, these results suggest the involvement of CB1R-MOR and CB1R-DOR complexes in diabetic neuropathy and that hemopressin could be developed as a potential therapeutic for the treatment of this painful condition.
Subject(s)
Cannabinoids , Diabetes Mellitus, Experimental , Diabetic Neuropathies , Neuralgia , Animals , Diabetes Mellitus, Experimental/complications , Diabetic Neuropathies/drug therapy , Disease Models, Animal , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Ligands , Mice , Neuralgia/drug therapy , Receptors, Opioid , Receptors, Opioid, mu/metabolismABSTRACT
ABSTRACT: Primary headache conditions are frequently associated with multiple sclerosis (MS), but the mechanism that triggers or worsens headaches in patients with MS is poorly understood. We previously showed that the proalgesic transient receptor potential ankyrin 1 (TRPA1) mediates hind paw mechanical and cold allodynia in a relapsing-remitting experimental autoimmune encephalomyelitis (RR-EAE) model in mice. Here, we investigated the development of periorbital mechanical allodynia (PMA) in RR-EAE, a hallmark of headache, and if TRPA1 contributed to this response. RR-EAE induction by injection of the myelin oligodendrocyte peptide fragment35-55 (MOG35-55) and Quillaja A adjuvant (Quil A) in C57BL/6J female mice elicited a delayed and sustained PMA. The PMA at day 35 after induction was reduced by the calcitonin gene-related peptide receptor antagonist (olcegepant) and the serotonin 5-HT1B/D receptor agonist (sumatriptan), 2 known antimigraine agents. Genetic deletion or pharmacological blockade of TRPA1 attenuated PMA associated with RR-EAE. The levels of oxidative stress biomarkers (4-hydroxynonenal and hydrogen peroxide, known TRPA1 endogenous agonists) and superoxide dismutase and NADPH oxidase activities were increased in the trigeminal ganglion of RR-EAE mice. Besides, the treatment with antioxidants (apocynin or α-lipoic acid) attenuated PMA. Thus, the results of this study indicate that TRPA1, presumably activated by endogenous agonists, evokes PMA in a mouse model of relapsing-remitting MS.
Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis, Relapsing-Remitting , Multiple Sclerosis , Animals , Ankyrins , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/complications , Female , Headache/complications , Hyperalgesia/complications , Hyperalgesia/etiology , Mice , Mice, Inbred C57BL , Multiple Sclerosis/complications , Multiple Sclerosis, Relapsing-Remitting/complications , Multiple Sclerosis, Relapsing-Remitting/drug therapy , TRPA1 Cation Channel/geneticsABSTRACT
ABSTRACT: IMT504, a noncoding, non-CpG oligodeoxynucleotide, modulates pain-like behavior in rats undergoing peripheral nerve injury, through mechanisms that remain poorly characterized. Here, we chose the spared nerve injury model in rats to analyze the contribution of mesenchymal stem cells (MSCs) in the mechanisms of action of IMT504. We show that a single subcutaneous administration of IMT504 reverses mechanical and cold allodynia for at least 5 weeks posttreatment. This event correlated with long-lasting increases in the percentage of MSCs in peripheral blood and injured sciatic nerves, in a process seemingly influenced by modifications in the CXCL12-CXCR4 axis. Also, injured nerves presented with reduced tumor necrosis factor-α and interleukin-1ß and increased transforming growth factor-ß1 and interleukin-10 protein levels. In vitro analysis of IMT504-pretreated rat or human MSCs revealed internalized oligodeoxynucleotide and confirmed its promigratory effects. Moreover, IMT504-pretreatment induced transcript expression of Tgf-ß1 and Il-10 in MSCs; the increase in Il-10 becoming more robust after exposure to injured nerves. Ex vivo exposure of injured nerves to IMT504-pretreated MSCs confirmed the proinflammatory to anti-inflammatory switch observed in vivo. Interestingly, the sole exposure of injured nerves to IMT504 also resulted in downregulated Tnf-α and Il-1ß transcripts. Altogether, we reveal for the first time a direct association between the antiallodynic actions of IMT504, its promigratory and cytokine secretion modulating effects on MSCs, and further anti-inflammatory actions at injured nerves. The recapitulation of key outcomes in human MSCs supports the translational potential of IMT504 as a novel treatment for neuropathic pain with a unique mechanism of action involving the regulation of neuroimmune interactions.
Subject(s)
Hyperalgesia , Mesenchymal Stem Cells , Animals , Anti-Inflammatory Agents , Hyperalgesia/etiology , Hyperalgesia/therapy , Interleukin-10 , Oligodeoxyribonucleotides/pharmacology , Rats , Rats, Sprague-Dawley , Sciatic Nerve/metabolism , Tumor Necrosis Factor-alpha/metabolismABSTRACT
This study aimed to investigate the central involvement of 5-HT1A receptors in the nociceptive behavior of mice submitted to the chronic constriction injury (CCI) of sciatic nerve and the subsequent application of photobiomodulation (PBM). Male mice (Swiss-albino) were submitted to CCI and subsequently received an infusion of WAY100635 (5-HT1A receptor antagonist) or intracerebroventricular saline (ICV), followed by infrared laser irradiation (808 nm), in continuous mode, with the power of 100 mW and a dose of 0 J/cm2 (control group) or 50 J/cm2. The thermal hyperalgesia was evaluated by hot plate test, while mechanical allodynia was evaluated by von Frey filaments. After CCI, animals showed a reduction in the nociceptive threshold (p<0.001) when compared to the sham group. In von Frey test, the CCI + saline + PBM 50 J/cm2 group showed an increase in nociceptive threshold (p<0.001) in all measurement moments in comparison with groups CCI + SALINE + PBM 0 J/cm2, CCI + WAY100635 + PBM 50 J/cm2, and CCI + WAY100635 + PBM 0 J/cm2. Similarly, in hot plate test, CCI + SALINE + PBM 50 J/cm2 group showed an increase in nociceptive threshold after application of PBM at 120 and 180 min. Because of the results found, it can be suggested the involvement of 5-HT1A receptors in the central nervous system, since WAY100635 was able to reverse the antinociceptive effect provided by PBM in animals submitted to CCI.
Subject(s)
Neuralgia , Receptor, Serotonin, 5-HT1A , Animals , Disease Models, Animal , Hyperalgesia/etiology , Hyperalgesia/radiotherapy , Male , Mice , Neuralgia/radiotherapy , Sciatic NerveABSTRACT
STUDY DESIGN: Prospective longitudinal experimental study. OBJECTIVE: We evaluate the effect of dapsone on tactile allodynia and mechanical hyperalgesia and to determine its anti-oxidant effect in a spinal cord injury (SC) model in rats. SUMMARY OF BACKGROUND DATA: Neuropathic pain (NP) as result of traumatic spinal cord injury is a deleterious medical condition with temporal or permanent time-course. Painful stimuli trigger a cascade of events that activate the N-methyl-D-aspartate (NMDA) receptor, inducing an increase in oxidative stress. Since there is no effective treatment for this condition, dapsone (4,4'diaminodiphenylsulfone) is proposed as potential treatment for NP. Its anti-oxidant, neuroprotective, and anti-inflammatory properties have been documented, however, there is no evidence regarding its use for treatment of NP induced by SCI. METHODS: In this study, we evaluated the anti-allodynic and anti-hyperalgesic effect of dapsone as preventive or acute treatment after NP was already established. Furthermore, participation of oxidative stress was evaluated by measuring lipid peroxidation (LP) and glutathione concentration (GSH) in rats with SCI. RESULTS: Acute treatment with dapsone (3.1-25âmg/kg, i.p.) decreased nociceptive behaviors in a dose-dependent manner, decreased LP, and increased GSH in the injured tissue 15âdays after the injury was produced. On the other hand, preventive treatment (3âh post-injury, once daily for 3âdays) with dapsone (3.1-25âmg/kg, i.p.) yielded similar results. CONCLUSION: The findings suggest that the anti-nociceptive effect of dapsone is regulated through the decrease of oxidative stress and the excitotoxicity is associated with the activation of NMDA receptors.Level of Evidence: N/A.
Subject(s)
Neuralgia , Spinal Cord Injuries , Animals , Dapsone/pharmacology , Disease Models, Animal , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Hyperalgesia/prevention & control , Oxidative Stress , Prospective Studies , Rats , Rats, Sprague-Dawley , Spinal Cord , Spinal Cord Injuries/complications , Spinal Cord Injuries/drug therapyABSTRACT
AIMS: Breast cancer-induced chronic pain is usually treated with opioids, but these compounds cause various adverse effects. Transient receptor potential ankyrin 1 (TRPA1) is involved in cancer pain; also, endogenous TRPA1 agonists are associated with cancer pain development. The aim of this study was to observe the antinociceptive effect of a repeated-dose TRPA1 antagonist administration and the production of endogenous TRPA1 agonists and TRPA1 expression in bone tissue in a model of breast cancer pain in mice. Second, we used a sequence reading archive (SRA) strategy to observe the presence of this channel in the mouse bone and in mouse bone cell lines. MAIN METHODS: We used BALB/c mice for experiments. The animals were subjected to the tumor cell inoculation (4 T1 strain). HC-030031 (a TRPA1 antagonist) treatment was done from day 11 to day 20 after tumor inoculation. TRPA1 expression and biochemical tests of oxidative stress were performed in the bone of mice (femur). SRA strategy was used to detect the TRPA1 presence. KEY FINDINGS: Repeated treatment with the TRPA1 antagonist produced an antinociceptive effect. There was an increase in hydrogen peroxide levels, NADPH oxidase and superoxide dismutase activities, but the expression of TRPA1 in the bone tissue was not altered. SRA did not show TRPA1 residual transcription in the osteoblast and osteoclast cell lines, as well as for mice cranial tissue and in mouse osteoclast precursors. SIGNIFICANCE: The TRPA1 receptor is a potential target for the development of new painkillers for the treatment of bone cancer pain.
Subject(s)
Acetanilides/pharmacology , Bone and Bones/drug effects , Cancer Pain/drug therapy , Hyperalgesia/drug therapy , Mammary Neoplasms, Animal/complications , Nociception/drug effects , Purines/pharmacology , TRPA1 Cation Channel/antagonists & inhibitors , Acetanilides/administration & dosage , Animals , Bone and Bones/metabolism , Cancer Pain/etiology , Cancer Pain/metabolism , Cancer Pain/pathology , Female , Hyperalgesia/etiology , Hyperalgesia/metabolism , Hyperalgesia/pathology , Mice , Mice, Inbred BALB C , Purines/administration & dosageABSTRACT
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 useABSTRACT
Agents that modulate the activity of high-voltage gated calcium channels (HVCCs) exhibit experimentally and clinically significant effect by relieving visceral pain. Among these agents, the toxins Phα1ß and ω-conotoxin MVIIA effectively reduce chronic pain in rodent models. The molecular mechanisms underlying the chronic pain associated with acute pancreatitis (AP) are poorly understood. Hypercalcemia is a risk factor; the role of cytosolic calcium is considered to be a modulator of pancreatitis. Blockade of Ca2+ signals may be useful as a prophylactic treatment of pancreatitis. We explored the pathophysiological roles of three peptide toxins: Phα1ß and its recombinant form CTK 01512-2-blockers of TRPA1 receptor and HVCCs and ω-conotoxin MVIIA, a specific blocker of N-type calcium channels in cerulein-induced AP. Cerulein injection elicits AP in rats, evidenced by an increase in hyperalgesic pain, inflammatory infiltration, amylase and lipase secretion, and reactive oxygen species, TNF-α, and p65 NF-κB levels. These effects of cerulein-induced AP were abolished by Phα1ß and its recombinant form CTK 01512-2, whereas ω-conotoxin MVIIA had no effect on the induced increase in pancreatic enzyme secretion. Our results demonstrate that Phα1ß and CTK 01512-2 toxins-antagonists of HVCCs and TRPA1 receptor presented an effective response profile, in the control of nociception and inflammatory process in the AP model in rats, without causing changes in spontaneous locomotion of the rats.
Subject(s)
Abdominal Pain/prevention & control , Analgesics/pharmacology , Anti-Inflammatory Agents/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Hyperalgesia/prevention & control , Pain Threshold/drug effects , Pancreatitis/prevention & control , Abdominal Pain/etiology , Abdominal Pain/metabolism , Abdominal Pain/physiopathology , Animals , Behavior, Animal/drug effects , Calcium Channels/metabolism , Calcium Signaling/drug effects , Ceruletide , Disease Models, Animal , Exploratory Behavior/drug effects , Hyperalgesia/etiology , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Inflammation Mediators/metabolism , Male , Neuropeptides/pharmacology , Pancreas/drug effects , Pancreas/metabolism , Pancreatitis/chemically induced , Pancreatitis/metabolism , Pancreatitis/physiopathology , Rats, Wistar , Spider Venoms/pharmacology , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/physiopathology , omega-Conotoxins/pharmacologyABSTRACT
Chronic pain affects significant portion of the world's population and physical exercise has been extensively indicated as non-pharmacological clinical intervention to relieve symptoms in chronic pain conditions. In general, studies on pain chronification and physical exercise intervention have focused on neuropathic pain, although chronic pain commonly results from an original inflammatory episode. Based on this, the objective of the present study was to investigate the therapeutic and preventive effect of the running wheel exercise on the persistent hyperalgesia induced by repetitive inflammatory stimulus, a rodent model that simulates clinical conditions of chronic pain that persist even with no more inflammatory stimulus present. To evaluate the therapeutic effect of physical exercise, we first induced persistent hyperalgesia through 14 days of PGE2 hind paw injections and, after that, mice have access to the regular voluntary running wheel. To evaluate the preventive effect of physical exercise, we first left the mice with access to the regular voluntary running wheel and, after that, we performed 14 days of PGE2 hind paw injection. Our results showed that voluntary running wheel exercise reduced persistent mechanical and chemical hyperalgesia intensity induced by repetitive inflammatory stimulus. In addition, we showed that this therapeutic effect is long-lasting and is observed even if started belatedly, i.e. two weeks after the development of hyperalgesia. Also, our results showed that voluntary running wheel exercise absolutely prevented persistent mechanical and chemical hyperalgesia induction. We can conclude that physical exercise has therapeutic and preventive effect on inflammatory stimulus-induced persistent hyperalgesia. Our data from animal experiments bypass placebo effects bias of the human studies and reinforce physical exercise clinical recommendations to treat and prevent chronic pain.
Subject(s)
Exercise Therapy , Hyperalgesia/etiology , Hyperalgesia/therapy , Inflammation/complications , Animals , Chronic Pain/etiology , Chronic Pain/prevention & control , Chronic Pain/therapy , Disease Models, Animal , Hyperalgesia/prevention & control , Male , Mice , Mice, Inbred C57BL , Physical Conditioning, Animal , RunningABSTRACT
The aim of this investigation was to evaluate the effects of local anaesthesia on nerve growth factor (NGF) induced masseter hyperalgesia. Healthy participants randomly received an injection into the right masseter muscle of either isotonic saline (IS) given as a single injection (n = 15) or an injection of NGF (n = 30) followed by a second injection of lidocaine (NGF + lidocaine; n = 15) or IS (NGF + IS; n = 15) in the same muscle 48 h later. Mechanical sensitivity scores of the right and left masseter, referred sensations and jaw pain intensity and jaw function were assessed at baseline, 48 h after the first injection, 5 min after the second injection and 72 h after the first injection. NGF caused significant jaw pain evoked by chewing at 48 and 72 h after the first injection when compared to the IS group, but without significant differences between the NGF + lidocaine and NGF + IS groups. However, the mechanical sensitivity of the right masseter 5 min after the second injection in the NGF + lidocaine group was significantly lower than the second injection in the NGF + IS and was similar to the IS group. There were no significant differences for the referred sensations. Local anaesthetics may provide relevant information regarding the contribution of peripheral mechanisms in the maintenance of persistent musculoskeletal pain.
Subject(s)
Anesthetics, Local/administration & dosage , Facial Pain/drug therapy , Hyperalgesia/drug therapy , Lidocaine/administration & dosage , Masseter Muscle/drug effects , Nerve Growth Factor/adverse effects , Adult , Case-Control Studies , Double-Blind Method , Facial Pain/etiology , Facial Pain/pathology , Female , Humans , Hyperalgesia/etiology , Hyperalgesia/pathology , Injections, Intramuscular , Male , Masseter Muscle/physiopathology , Pain ThresholdABSTRACT
Acute pain that persists for a few days is associated with a reduction in patients' quality of life. Orofacial persistent pain promotes psychological disorders such as anxiety, impairs daily essential activities such as eating, and results in decreased social interaction. Here, we investigated whether rats subjected to orofacial formalin injection or intraoral incision surgery display persistent facial heat hyperalgesia, ongoing pain, anxiety-like behavior, and changes in ultrasonic vocalization. Orofacial formalin injection or intraoral incision caused facial heat hyperalgesia for 3 days compared with saline-injected and sham animals. In addition, both experimental groups showed a reduction in the number of entries and in the time spent in the open arms in the elevated plus maze test on day 3, suggesting that anxiety-like behavior developed as a consequence of persistent pain. At this time point, both groups also displayed a reduction in the number of 50-kHz calls, specifically in the flat subtype, which suggests a decrease in social communication. Moreover, on day 3 after surgery, systemic morphine produced robust conditioned place preference in rats subjected to intraoral incision compared with sham, and the former group also presented increased spontaneous facial grooming, revealing the presence of ongoing pain. Finally, Western blot and immunohistochemistry analysis showed a reduction in tyrosine hydroxylase expression in the nucleus accumbens, which may reflect a decrease in mesolimbic dopaminergic activity. Altogether, the results demonstrate that acute orofacial pain causes prolonged changes in behavioral and affective pain components, which may be related to dopaminergic changes in the nucleus accumbens.
Subject(s)
Acute Pain , Animals , Disease Models, Animal , Facial Pain , Humans , Hyperalgesia/etiology , Quality of Life , Rats , Rats, WistarABSTRACT
ETHNOPHARMACOLOGY RELEVANCE: Schinus terebinthifolia Raddi leaves have been used in folk medicine due to several properties, including antitumor and analgesic effects. The variable efficacy and adverse effects of analgesic drugs have motivated the search for novel antinociceptive agents. It has been reported that the S. terebinthifolia leaf lectin (SteLL) has antitumor activity against sarcoma 180 in mice. AIM OF THE STUDY: This work aimed to evaluate whether SteLL would reduce cancer pain using an orthotopic tumor model. MATERIALS AND METHODS: A sarcoma 180 cell suspension was inoculated into the right hind paws of mice, and the treatments (150 mM NaCl, negative control; 10 mg/kg morphine, positive control; or SteLL at 1 and 2 mg/kg) were administered intraperitoneally 24 h after cell inoculation up to 14 days. Spontaneous nociception, mechanical hyperalgesia, and hot-plate tests were performed. Further, the volume and weight of the tumor-bearing paws were measured. RESULTS: SteLL (2 mg/kg) improved limb use during ambulation. The lectin (1 and 2 mg/kg) also inhibited mechanical hyperalgesia and increased the latency time during the hot-plate test. Naloxone was found to reverse this effect, indicating the involvement of opioid receptors. The tumor-bearing paws of mice treated with SteLL exhibited lower volume and weight. CONCLUSION: SteLL reduced hyperalgesia due to sarcoma 180 in the paws of mice, and this effect can be related to its antitumor action.
Subject(s)
Anacardiaceae , Analgesics/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cancer Pain/prevention & control , Hyperalgesia/prevention & control , Nociceptive Pain/prevention & control , Plant Leaves , Plant Lectins/pharmacology , Sarcoma 180/drug therapy , Anacardiaceae/chemistry , Analgesics/isolation & purification , Animals , Antineoplastic Agents, Phytogenic/isolation & purification , Cancer Pain/etiology , Cancer Pain/metabolism , Cancer Pain/physiopathology , Female , Hyperalgesia/etiology , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Mice , Nociception/drug effects , Nociceptive Pain/etiology , Nociceptive Pain/metabolism , Nociceptive Pain/physiopathology , Pain Threshold/drug effects , Plant Leaves/chemistry , Plant Lectins/isolation & purification , Reaction Time/drug effects , Receptors, Opioid/metabolism , Sarcoma 180/complications , Sarcoma 180/pathology , Signal Transduction , Time FactorsABSTRACT
BACKGROUND AND OBJECTIVES: Complex regional pain syndrome (CRPS) is defined as an extreme and chronic pain condition, and photobiomodulation has relevance as a complementary treatment for CRPS. The objective of this study was to verify the effects of photobiomodulation (PBMT) therapy protocols at two wavelengths 660 and 830 nm, associated or not to nicotine in complex regional pain syndrome type I (CRPS-I). STUDY DESIGN/MATERIALS AND METHODS: Sixty-four Swiss mice were divided into the following groups: (i) Naive, (ii) Sham, (iii) Control, (iv) 660 nm, (v) 830 nm, (vii) Nicotine, (vii) Nicotine/660 nm, and (viii) Nicotine/830 nm. CRPS-I was induced in an experimental ischemia/reperfusion model by affixing an elastic ring, proximal to the ankle joint of the right hind mouse paw, for 3 hours. Nicotine, in the respective groups was administered for 28 days prior to the induction of CRPS-I. PBMT was applied immediately after the procedure and for 20 consecutive days. The animals were evaluated for mechanical hyperalgesia, thermal hyperalgesia, paw edema at baseline and for 7, 14, and 21 days. Statistical analyses comprised a mixed-effects model, using the Tukey post hoc test (P < 0.05). RESULTS: The PBMT wavelengths in 660 and 830 nm groups had beneficial effects (P < 0.05) in reducing mechanical and thermal hyperalgesia, but the effects at 660 nm were significantly better than 830 nm. At reducing edema, both wavelengths had significant effects statistically, absolutely no difference between them. CONCLUSIONS: The use of PBMT (660 and 830 nm) was effective in reducing mechanical hyperalgesia and thermal hyperalgesia; however, PBMT at 660 nm generated significant results. In reducing edema, both wavelengths had similar effects, which were significant statistically. The deleterious effects of nicotine were evident statistically and were softened when treated with PBMT (P < 0.05). Lasers Surg. Med. © 2020 Wiley Periodicals, Inc.
Subject(s)
Chronic Pain , Complex Regional Pain Syndromes , Animals , Complex Regional Pain Syndromes/therapy , Disease Models, Animal , Hyperalgesia/etiology , Mice , NicotineABSTRACT
Central neuropathic pain is a common untreated symptom in progressive multiple sclerosis (PMS) and is associated with poor quality of life and interference with patients' daily activities. The neuroinflammation process and mitochondrial dysfunction in the PMS lesions generate reactive species. The transient potential receptor ankyrin 1 (TRPA1) has been identified as one of the major mechanisms that contribute to neuropathic pain signaling and can be activated by reactive compounds. Thus, the goal of our study was to evaluate the role of spinal TRPA1 in the central neuropathic pain observed in a PMS model in mice. We used C57BL/6 female mice (20-30 g), and the PMS model was induced by the experimental autoimmune encephalomyelitis (EAE) using mouse myelin oligodendrocyte glycoprotein (MOG35-55) antigen and CFA (complete Freund's adjuvant). Mice developed progressive clinical score, with motor impairment observed after 15 days of induction. This model induced mechanical and cold allodynia and heat hyperalgesia which were measured up to 14 days after induction. The hypersensitivity observed was reduced by the administration of selective TRPA1 antagonists (HC-030031 and A-967079, via intrathecal and intragastric), antioxidants (α-lipoic acid and apocynin, via intrathecal and intragastric), and TRPA1 antisense oligonucleotide (via intrathecal). We also observed an increase in TRPA1 mRNA levels, NADPH oxidase activity, and 4-hydroxinonenal (a TRPA1 agonist) levels in spinal cord samples of PMS-EAE induced animals. In conclusion, these results support the hypothesis of the TRPA1 receptor involvement in nociception observed in a PMS-EAE model in mice.
Subject(s)
Encephalomyelitis, Autoimmune, Experimental/complications , Hyperalgesia/physiopathology , Nerve Tissue Proteins/physiology , Neuralgia/physiopathology , Nociception/physiology , Spinal Cord/physiopathology , TRPA1 Cation Channel/physiology , Acetanilides/pharmacology , Acetanilides/therapeutic use , Acetophenones/pharmacology , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Antipyrine/analogs & derivatives , Antipyrine/pharmacology , Antipyrine/therapeutic use , Dipyrone/pharmacology , Dipyrone/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein/toxicity , NADPH Oxidases/antagonists & inhibitors , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuralgia/drug therapy , Neuralgia/etiology , Nociception/drug effects , Oligonucleotides, Antisense/pharmacology , Oxidative Stress , Oximes/pharmacology , Oximes/therapeutic use , Peptide Fragments/immunology , Peptide Fragments/toxicity , Pregabalin/pharmacology , Pregabalin/therapeutic use , Purines/pharmacology , Purines/therapeutic use , TRPA1 Cation Channel/antagonists & inhibitors , TRPA1 Cation Channel/biosynthesis , TRPA1 Cation Channel/genetics , Thioctic Acid/pharmacology , Up-Regulation/drug effectsABSTRACT
Central neuropathic pain is the main symptom caused by spinal cord lesion in relapsing-remitting multiple sclerosis (RRMS), but its management is still not effective. The transient receptor potential ankyrin 1 (TRPA1) is a pain detecting ion channel involved in neuropathic pain development. Thus, the aim of our study was to evaluate the role of TRPA1 in central neuropathic nociception induced by relapsing-remitting experimental autoimmune encephalomyelitis (RR-EAE) mouse model. In this model, we observed the development of similar clinical conditions of RRMS in C57BL/6 female mice through RR-EAE using MOG35-55 antigen and Quil A adjuvant. At the thirty-fifth day post-induction, C57BL/6 female mice demonstrated alteration in the RR-EAE score without motor impairment, mechanical and cold allodynia. Also, significative changes in demyelinating (Mog and olig-1) and neuroinflammatory (Iba1, Gfap and Tnfa) markers were observed, but this model did not alter Trpa1 RNA expression levels in the spinal cord. The hydrogen peroxide and 4-hydroxynonenal levels (TRPA1 agonists) were increased in RR-EAE induced mice, as well as the NADPH oxidase activity. The intragastric treatment of RR-EAE induced mice with TRPA1 antagonists (HC-030031 and A-967079) and antioxidant (α-lipoic acid and apocynin) caused an antiallodynic effect. Moreover, the intrathecal administration of TRPA1 antisense oligonucleotide, HC-030031, α-lipoic acid, and apocynin transiently attenuated mechanical and cold allodynia. Thus, TRPA1 plays a key role in the induction of neuropathic pain in this model of RR-EAE and can be a possible target for investigating the development of pain in RRMS patients.