24 results on '"Popiolek-Barczyk K"'
Search Results
2. Involvement of microglial cells in the antinociceptive effects of metamizol in a mouse model of neuropathic pain
- Author
-
Zajaczkowska, R., primary, Popiolek-Barczyk, K., additional, Pilat, D., additional, Rojewska, E., additional, Makuch, W., additional, Wordliczek, J., additional, and Mika, J., additional
- Published
- 2018
- Full Text
- View/download PDF
3. Long pentraxin PTX3 is upregulated systemically and centrally after experimental neurotrauma, but its depletion leaves unaltered sensorimotor deficits or histopathology.
- Author
-
Oggioni M, Mercurio D, Minuta D, Fumagalli S, Popiolek-Barczyk K, Sironi M, Ciechanowska A, Ippati S, De Blasio D, Perego C, Mika J, Garlanda C, and De Simoni MG
- Subjects
- Animals, Brain Injuries genetics, Brain Injuries pathology, C-Reactive Protein genetics, Collagen metabolism, Disease Models, Animal, Inflammation metabolism, Inflammation pathology, Male, Mice, Neurons pathology, Neutrophils metabolism, Serum Amyloid P-Component genetics, Brain metabolism, Brain Injuries metabolism, C-Reactive Protein metabolism, Neurons metabolism, Serum Amyloid P-Component metabolism, Up-Regulation
- Abstract
Long pentraxin PTX3, a pattern recognition molecule involved in innate immune responses, is upregulated by pro-inflammatory stimuli, contributors to secondary damage in traumatic brain injury (TBI). We analyzed PTX3 involvement in mice subjected to controlled cortical impact, a clinically relevant TBI mouse model. We measured PTX3 mRNA and protein in the brain and its circulating levels at different time point post-injury, and assessed behavioral deficits and brain damage progression in PTX3 KO mice. PTX3 circulating levels significantly increased 1-3 weeks after injury. In the brain, PTX3 mRNA was upregulated in different brain areas starting from 24 h and up to 5 weeks post-injury. PTX3 protein significantly increased in the brain cortex up to 3 weeks post-injury. Immunohistochemical analysis showed that, 48 h after TBI, PTX3 was localized in proximity of neutrophils, likely on neutrophils extracellular traps (NETs), while 1- and 2- weeks post-injury PTX3 co-localized with fibrin deposits. Genetic depletion of PTX3 did not affect sensorimotor deficits up to 5 weeks post-injury. At this time-point lesion volume and neuronal count, axonal damage, collagen deposition, astrogliosis, microglia activation and phagocytosis were not different in KO compared to WT mice. Members of the long pentraxin family, neuronal pentraxin 1 (nPTX1) and pentraxin 4 (PTX4) were also over-expressed in the traumatized brain, but not neuronal pentraxin 2 (nPTX2) or short pentraxins C-reactive protein (CRP) and serum amyloid P-component (SAP). The long-lasting pattern of activation of PTX3 in brain and blood supports its specific involvement in TBI. The lack of a clear-cut phenotype in PTX3 KO mice may depend on the different roles of this protein, possibly involved in inflammation early after injury and in repair processes later on, suggesting distinct functions in acute phases versus sub-acute or chronic phases. Brain long pentraxins, such as PTX4-shown here to be overexpressed in the brain after TBI-may compensate for PTX3 absence.
- Published
- 2021
- Full Text
- View/download PDF
4. The CCL2/CCL7/CCL12/CCR2 pathway is substantially and persistently upregulated in mice after traumatic brain injury, and CCL2 modulates the complement system in microglia.
- Author
-
Popiolek-Barczyk K, Ciechanowska A, Ciapała K, Pawlik K, Oggioni M, Mercurio D, De Simoni MG, and Mika J
- Subjects
- Animals, Brain metabolism, Brain pathology, Cells, Cultured, Chemokine CCL2 genetics, Chemokine CCL7 genetics, Inflammation Mediators metabolism, Lipopolysaccharides, Male, Mice, Inbred C57BL, Monocyte Chemoattractant Proteins genetics, RNA, Messenger genetics, RNA, Messenger metabolism, Receptors, CCR2 genetics, Signal Transduction, Time Factors, Brain Injuries, Traumatic genetics, Chemokine CCL2 metabolism, Chemokine CCL7 metabolism, Complement System Proteins metabolism, Microglia metabolism, Monocyte Chemoattractant Proteins metabolism, Receptors, CCR2 metabolism, Up-Regulation
- Abstract
Traumatic brain injury (TBI) is the leading cause of death in the global population. Disturbed inflammatory processes after TBI exacerbate secondary brain injury and contribute to unfavorable outcomes. Multiple inflammatory events that accompany brain trauma, such as glial activation, chemokine release, or the initiation of the complement system cascade, have been identified as potential targets for TBI treatment. However, the participation of chemokines in the complement activation remains unknown. Our studies sought to determine the changes in the expression of the molecules involved in the CCL2/CCL7/CCL12/CCR2 pathway in the injured brain and the effect of CCL2, CCL7, and CCL12 (10, 100, and 500 ng/mL) on the classic and lectin complement pathways and inflammatory factors in microglial cell cultures. Brain injury in mice was modeled by controlled cortical impact (CCI). Our findings indicate a time-dependent upregulation of CCL2, CCL7, and CCL12 at the mRNA and protein levels within the cortex, striatum, and/or thalamus beginning 24 h after the trauma. The analysis of the expression of the receptor of the tested chemokines, CCR2, revealed its substantial upregulation within the injured brain areas mainly on the mRNA level. Using primary cortical microglial cell cultures, we observed a substantial increase in the expression of CCL2, CCL7, and CCL12 after 24 h of LPS (100 ng/mL) treatment. CCL2 stimulation of microglia increased the level of IL-1β mRNA but did not influence the expression of IL-18, IL-6, and IL-10. Moreover, CCL2 significantly increased the expression of Iba1, a marker of microglia activation. CCL2 and CCL12 upregulated the expression of C1qa but did not influence the expression of C1ra and C1s1 (classical pathway); moreover, CCL2 increased ficolin A expression and reduced collectin 11 expression (lectin pathway). Additionally, we observed the downregulation of pentraxin 3, a modulator of the complement cascade, after CCL2 and CCL12 treatment. We did not detect the expression of ficolin B, Mbl1, and Mbl2 in microglial cells. Our data identify CCL2 as a modulator of the classical and lectin complement pathways suggesting that CCL2 may be a promising target for pharmacological intervention after brain injury. Moreover, our study provides evidence that CCL2 and two other CCR2 ligands may play a role in the development of changes in TBI., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
5. Traumatic brain injury in mice induces changes in the expression of the XCL1/XCR1 and XCL1/ITGA9 axes.
- Author
-
Ciechanowska A, Popiolek-Barczyk K, Ciapała K, Pawlik K, Oggioni M, Mercurio D, de Simoni MG, and Mika J
- Subjects
- Animals, Astrocytes metabolism, Chemokines, C genetics, Disease Models, Animal, Disease Progression, Integrin alpha Chains genetics, Male, Mice, Mice, Inbred C57BL, Microglia metabolism, Brain Injuries, Traumatic physiopathology, Chemokines, C metabolism, Integrin alpha Chains metabolism, Receptors, Chemokine metabolism
- Abstract
Background: Every year, millions of people suffer from various forms of traumatic brain injury (TBI), and new approaches with therapeutic potential are required. Although chemokines are known to be involved in brain injury, the importance of X-C motif chemokine ligand 1 (XCL1) and its receptors, X-C motif chemokine receptor 1 (XCR1) and alpha-9 integrin (ITGA9), in the progression of TBI remain unknown., Methods: Using RT-qPCR/Western blot/ELISA techniques, changes in the mRNA/protein levels of XCL1 and its two receptors, in brain areas at different time points were measured in a mouse model of TBI. Moreover, their cellular origin and possible changes in expression were evaluated in primary glial cell cultures., Results: Studies revealed the spatiotemporal upregulation of the mRNA expression of XCL1, XCR1 and ITGA9 in all the examined brain areas (cortex, thalamus, and hippocampus) and at most of the evaluated stages after brain injury (24 h; 4, 7 days; 2, 5 weeks), except for ITGA9 in the thalamus. Moreover, changes in XCL1 protein levels occurred in all the studied brain structures; the strongest upregulation was observed 24 h after trauma. Our in vitro experiments proved that primary murine microglial and astroglial cells expressed XCR1 and ITGA9, however they seemed not to be a main source of XCL1., Conclusions: These findings indicate that the XCL1/XCR1 and XCL1/ITGA9 axes may participate in the development of TBI. The XCL1 can be considered as one of the triggers of secondary injury, therefore XCR1 and ITGA9 may be important targets for pharmacological intervention after traumatic brain injury.
- Published
- 2020
- Full Text
- View/download PDF
6. CCR4 antagonist (C021) influences the level of nociceptive factors and enhances the analgesic potency of morphine in a rat model of neuropathic pain.
- Author
-
Bogacka J, Popiolek-Barczyk K, Pawlik K, Ciechanowska A, Makuch W, Rojewska E, Dobrogowski J, Przeklasa-Muszynska A, and Mika J
- Subjects
- Animals, Cold Temperature, Cytokines genetics, Cytokines metabolism, Disease Models, Animal, Drug Synergism, Ganglia, Spinal drug effects, Ganglia, Spinal metabolism, Male, Mice, Rats, Wistar, Receptors, CCR4 genetics, Sciatic Nerve injuries, Spinal Cord drug effects, Spinal Cord metabolism, Touch, Analgesics, Opioid administration & dosage, Hyperalgesia drug therapy, Morphine administration & dosage, Neuralgia drug therapy, Peripheral Nerve Injuries drug therapy, Quinazolines administration & dosage, Receptors, CCR4 antagonists & inhibitors
- Abstract
Neuropathic pain is a chronic condition which significantly reduces the quality of life and serious clinical issue that is in general resistant to available therapies. Therefore looking for new analgesics is still critical issue. Recent, studies have indicated that chemokine signaling pathways are crucial for the development of neuropathy; however, the role of CC chemokine receptor 4 (CCR4) in this process has not yet been studied. Therefore, the aim of our research was to investigate the influence of C021 (a CCR4 antagonist) and CCR4 CC chemokine ligands 17 and 22 (CCL17 and CCL22) on the development of hypersensitivity and the effectiveness of morphine induced analgesia in naive animals and/or animals exposed to chronic constriction injury (CCI) of the sciatic nerve. Firstly, we demonstrated that the intrathecal administration of CCL17 and CCL22 induced pain-related behavior in naive mice. Secondly, we revealed that the intrathecal injection of C021 significantly reduced CCI-induced hypersensitivity after nerve injury. In parallel, C021 reduced microglia/macrophages activation and the level of some pronociceptive interleukins (IL-1beta; IL-18) in the spinal cord 8 days after CCI. Moreover, C021 not only attenuated mechanical and thermal hypersensitivity but also enhanced the analgesic properties of morphine. Our research indicates that CCR4 ligands might be important factors in the early stages of neuropathy, when we observe intense microglia/macrophages activation. Moreover, pharmacological blockade of CCR4 may serve as a potential new target for better understanding the mechanisms of neuropathic pain development., Competing Interests: Declaration of competing interest The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2020 Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
7. Changes in macrophage inflammatory protein-1 (MIP-1) family members expression induced by traumatic brain injury in mice.
- Author
-
Ciechanowska A, Popiolek-Barczyk K, Pawlik K, Ciapała K, Oggioni M, Mercurio D, De Simoni MG, and Mika J
- Subjects
- Animals, Biomarkers, Brain metabolism, Brain pathology, Brain Injuries, Traumatic pathology, Disease Models, Animal, Disease Susceptibility, Gene Expression Profiling, Macrophage Inflammatory Proteins metabolism, Mice, Microglia metabolism, Neurons metabolism, Neutrophils immunology, Neutrophils metabolism, Brain Injuries, Traumatic etiology, Brain Injuries, Traumatic metabolism, Gene Expression Regulation, Macrophage Inflammatory Proteins genetics, Multigene Family
- Abstract
A deep knowledge of the profound immunological response induced by traumatic brain injury (TBI) raises the possibility of novel therapeutic interventions. Existing studies have highlighted the important roles of C-C motif ligands in the development of neuroinflammation after brain injury; however, the participation of macrophage inflammatory protein-1 (MIP-1) family members in this phenomenon is still undefined. Therefore, the goal of our study was to evaluate changes in macrophage inflammatory protein-1 (MIP-1) family members (CCL3, CCL4, and CCL9) and their receptors (CCR1 and CCR5) in a mouse model of TBI (induced by controlled cortical impact (CCI)). We also investigated the pattern of activation of immunological cells (such as neutrophils, microglia and astroglia), which on one hand express CCR1/CCR5, and on the other hand might be a source of the tested chemokines in the injured brain. We investigated changes in mRNA (RT-qPCR) and/or protein (ELISA and Western blot) expression in brain structures (the cortex, hippocampus, thalamus, and striatum) at different time points (24 h, 4 days, 7 days, 2 weeks, and/or 5 weeks) after trauma. Our time-course studies revealed the upregulation of the mRNA expression of all members of the MIP-1 family (CCL3, CCL4, and CCL9) in all tested brain structures, mainly in the early stages after injury. A similar pattern of activation was observed at the protein level in the cortex and thalamus, where the strongest activation was observed 1 day after CCI; however, we did not observe any change in CCL3 in the thalamus. Analyses of CCR1 and CCR5 demonstrated the upregulation of the mRNA expression of both receptors in all tested cerebral structures, mainly in the early phases post injury (24 h, 4 days and 7 days). Protein analysis showed the upregulation of CCR1 and CCR5 in the thalamus 24 h after TBI, but we did not detect any change in the cortex. We also observed the upregulation of neutrophil marker (MPO) at the early time points (24 h and 7 days) in the cortex, while the profound activation of microglia (IBA-1) and astroglia (GFAP) was observed mainly on day 7. Our findings highlight for the first time that CCL3, CCL4, CCL9 and their receptors offer promising targets for influencing secondary neuronal injury and improving TBI therapy. The results suggest that the MIP-1 family is an important target for pharmacological intervention for brain injury., Competing Interests: Declaration of Competing Interest None., (Copyright © 2020 Elsevier GmbH. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
8. The blockade of CC chemokine receptor type 1 influences the level of nociceptive factors and enhances opioid analgesic potency in a rat model of neuropathic pain.
- Author
-
Pawlik K, Piotrowska A, Kwiatkowski K, Ciapała K, Popiolek-Barczyk K, Makuch W, and Mika J
- Subjects
- Animals, Calcium-Binding Proteins genetics, Calcium-Binding Proteins immunology, Chemokine CCL2 genetics, Chemokine CCL2 immunology, Disease Models, Animal, Drug Synergism, Ganglia, Spinal drug effects, Ganglia, Spinal immunology, Ganglia, Spinal physiopathology, Gene Expression Regulation, Hyperalgesia genetics, Hyperalgesia immunology, Hyperalgesia physiopathology, Interleukin-18 genetics, Interleukin-18 immunology, Interleukin-1beta genetics, Interleukin-1beta immunology, Interleukin-6 genetics, Interleukin-6 immunology, Male, Microfilament Proteins genetics, Microfilament Proteins immunology, Neuralgia genetics, Neuralgia immunology, Neuralgia physiopathology, Nociception drug effects, Peroxidase genetics, Peroxidase immunology, Protein Isoforms genetics, Protein Isoforms immunology, Rats, Rats, Wistar, Receptors, CCR1 antagonists & inhibitors, Receptors, CCR1 genetics, Sciatic Nerve drug effects, Sciatic Nerve injuries, Sciatic Nerve physiopathology, Signal Transduction, Analgesics pharmacology, Buprenorphine pharmacology, Hyperalgesia drug therapy, Morphine pharmacology, Neuralgia drug therapy, Receptors, CCR1 immunology, Xanthenes pharmacology
- Abstract
A growing body of evidence has indicated that the release of nociceptive factors, such as interleukins and chemokines, by activated immune and glial cells has crucial significance for neuropathic pain generation and maintenance. Moreover, changes in the production of nociceptive immune factors are associated with low opioid efficacy in the treatment of neuropathy. Recently, it has been suggested that CC chemokine receptor type 1 (CCR1) signaling is important for nociception. Our study provides evidence that the development of hypersensitivity in rats following chronic constriction injury (CCI) of the sciatic nerve is associated with significant up-regulation of endogenous CCR1 ligands, namely, CCL2, CCL3, CCL4, CCL6, CCL7 and CCL9 in the spinal cord and CCL2, CCL6, CCL7 and CCL9 in dorsal root ganglia (DRG). We showed that single and repeated intrathecal administration of J113863 (an antagonist of CCR1) attenuated mechanical and thermal hypersensitivity. Moreover, repeated administration of a CCR1 antagonist enhanced the analgesic properties of morphine and buprenorphine after CCI. Simultaneously, repeated administration of J113863 reduced the protein levels of IBA-1 in the spinal cord and MPO and CD4 in the DRG and, as a consequence, the level of pronociceptive factors, such as interleukin-1β (IL-1β), IL-6 and IL-18. The data obtained provide evidence that CCR1 blockade reduces hypersensitivity and increases opioid-induced analgesia through the modulation of neuroimmune interactions., (© 2020 John Wiley & Sons Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
9. Chemokines CCL2 and CCL7, but not CCL12, play a significant role in the development of pain-related behavior and opioid-induced analgesia.
- Author
-
Kwiatkowski K, Popiolek-Barczyk K, Piotrowska A, Rojewska E, Ciapała K, Makuch W, and Mika J
- Subjects
- Analgesia methods, Animals, Astrocytes metabolism, Cells, Cultured, Male, Mice, Microglia metabolism, Monocyte Chemoattractant Proteins metabolism, Neuroglia metabolism, Peripheral Nerve Injuries metabolism, Rats, Rats, Wistar, Sciatic Nerve metabolism, Spinal Cord metabolism, Analgesics, Opioid pharmacology, Chemokine CCL2 metabolism, Chemokine CCL7 metabolism, Hyperalgesia chemically induced, Hyperalgesia metabolism, Neuralgia chemically induced, Neuralgia metabolism
- Abstract
The complex neuroimmunological interactions mediated by chemokines are suggested to be responsible for the development of neuropathic pain. The lack of knowledge regarding the detailed pathomechanism of neuropathy is one reason for the lack of optimally efficient therapies. Recently, several lines of evidence indicated that expression of CCR2 is increased in spinal cord neurons and microglial cells after peripheral nerve injury. It was previously shown that administration of CCR2 antagonists induces analgesic effects; however, the role of CCR2 ligands in neuropathic pain still needs to be explained. Thus, the goal of our studies was to investigate the roles of CCL2, CCL7, and CCL12 in neuropathic pain development and opioid effectiveness. The experiments were conducted on primary glial cell cultures and two groups of mice: naive and neuropathic. We used chronic constriction injury (CCI) of the sciatic nerve as a neuropathic pain model. Mice intrathecally received chemokines (CCL2, CCL7, CCL12) at a dose of 10, 100 or 500 ng, neutralizing antibodies (anti-CCL2, anti-CCL7) at a dose of 1, 4 or 8 μg, and opioids (morphine, buprenorphine) at a dose of 1 μg. The pain-related behaviors were assessed using the von Frey and cold plate tests. The biochemical analysis of mRNA expression of glial markers, CCL2, CCL7 and CCL12 was performed using quantitative reverse transcriptase real-time PCR. We demonstrated that CCI of the sciatic nerve elevated spinal expression of CCL2, CCL7 and CCL12 in mice, in parallel with microglia and astroglial activation markers. Moreover, intrathecal injection of CCL2 and CCL7 induced pain-related behavior in naive mice in a dose-dependent manner. Surprisingly, intrathecal injection of CCL12 did not influence nociceptive transmission in naive or neuropathic mice. Additionally, we showed for the first time that intrathecal injection of CCL2 and CCL7 neutralizing antibodies not only attenuated CCI-induced pain-related behaviors in mice but also augmented the analgesia induced by morphine and buprenorphine. In vitro studies suggest that both microglia and astrocytes are an important cellular sources of the examined chemokines. Our results revealed the crucial roles of CCL2 and CCL7, but not CCL12, in neuropathic pain development and indicated that pharmacological modulation of these factors may serve as a potential therapeutic target for new (co)analgesics., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
10. Antinociceptive effects of novel histamine H 3 and H 4 receptor antagonists and their influence on morphine analgesia of neuropathic pain in the mouse.
- Author
-
Popiolek-Barczyk K, Łażewska D, Latacz G, Olejarz A, Makuch W, Stark H, Kieć-Kononowicz K, and Mika J
- Subjects
- Analgesia, Animals, Drug Synergism, Drug Therapy, Combination, Female, Male, Mice, Neuralgia physiopathology, Receptors, Histamine H4 physiology, Sciatic Nerve injuries, Analgesics, Opioid therapeutic use, Histamine Antagonists therapeutic use, Morphine therapeutic use, Neuralgia drug therapy, Receptors, Histamine H3 physiology, Receptors, Histamine H4 antagonists & inhibitors
- Abstract
Background and Purpose: The histaminergic system is a promising target for the development of new analgesics, as histamine H
3 and H4 receptors are expressed in regions concerned with nociceptive transmission. Here we have determined the analgesic effects of new H3 and H4 receptor antagonists in naive and neuropathic mice., Experimental Approach: We used chronic constriction injury (CCI) to the sciatic nerve in mice to model neuropathy. Effects of a new H3 receptor antagonist, E-162(1-(5-(naphthalen-1-yloxy)pentyl)piperidine) and H4 receptor antagonist, TR-7(4-(4-chlorophenyl)-6-(4-methylpiperazin-1-yl)-1,3,5-triazin-2-amine) were assessed on mechanical (von Frey) and thermal (cold plate, tail flick) stimuli in mice with and without CCI (7 days after injury). Effects of these antagonists on morphine analgesia were also evaluated, along with the possible participation of H1 receptors in their effects. We analysed the compounds in binding and functional cAMP assays at the H3 and H4 receptors and determined metabolic stability., Key Results: E-162 and TR-7 attenuated nociceptive responses and profound morphine analgesia in males with CCI. These antagonists showed analgesia in naive mice (tail flick test) and produced prolonged analgesia in neuropathic females. E-162-induced analgesia was reversed by pyrilamine, an H1 receptor antagonist. E-162 bound potently to H3 receptors (Ki = 55 nM) and inhibited cAMP accumulation (IC50 = 165 nM). TR-7 showed lower affinity for H4 receptors (Ki = 203 nM) and IC50 of 512 nM., Conclusions and Implications: We describe a therapeutic use for new H3 (E-162) and H4 receptor (TR-7) antagonists in neuropathy. Targeting H3 and H4 receptors enhanced morphine analgesia, consistent with multimodal pain therapy., (© 2018 The British Pharmacological Society.)- Published
- 2018
- Full Text
- View/download PDF
11. Botulinum Toxin Type A-A Modulator of Spinal Neuron-Glia Interactions under Neuropathic Pain Conditions.
- Author
-
Rojewska E, Piotrowska A, Popiolek-Barczyk K, and Mika J
- Subjects
- Animals, Humans, Neuroglia physiology, Neurons physiology, Spine cytology, Analgesics pharmacology, Analgesics therapeutic use, Botulinum Toxins, Type A pharmacology, Botulinum Toxins, Type A therapeutic use, Neuralgia drug therapy, Neuroglia drug effects, Neurons drug effects
- Abstract
Neuropathic pain represents a significant clinical problem because it is a chronic condition often refractory to available therapy. Therefore, there is still a strong need for new analgesics. Botulinum neurotoxin A (BoNT/A) is used to treat a variety of clinical diseases associated with pain. Glia are in continuous bi-directional communication with neurons to direct the formation and refinement of synaptic connectivity. This review addresses the effects of BoNT/A on the relationship between glia and neurons under neuropathic pain. The inhibitory action of BoNT/A on synaptic vesicle fusion that blocks the release of miscellaneous pain-related neurotransmitters is known. However, increasing evidence suggests that the analgesic effect of BoNT/A is mediated through neurons and glial cells, especially microglia. In vitro studies provide evidence that BoNT/A exerts its anti-inflammatory effect by diminishing NF-κB, p38 and ERK1/2 phosphorylation in microglia and directly interacts with Toll-like receptor 2 (TLR2). Furthermore, BoNT/A appears to have no more than a slight effect on astroglia. The full activation of TLR2 in astroglia appears to require the presence of functional TLR4 in microglia, emphasizing the significant interaction between those cell types. In this review, we discuss whether and how BoNT/A affects the spinal neuron-glia interaction and reduces the development of neuropathy., Competing Interests: The authors declare no conflict of interest.
- Published
- 2018
- Full Text
- View/download PDF
12. Analgesic Properties of Opioid/NK1 Multitarget Ligands with Distinct in Vitro Profiles in Naive and Chronic Constriction Injury Mice.
- Author
-
Starnowska J, Costante R, Guillemyn K, Popiolek-Barczyk K, Chung NN, Lemieux C, Keresztes A, Van Duppen J, Mollica A, Streicher J, Vanden Broeck J, Schiller PW, Tourwé D, Mika J, Ballet S, and Przewlocka B
- Subjects
- Animals, Chronic Disease, Constriction, Mice, Neuralgia drug therapy, Receptors, Neurokinin-1 drug effects, Receptors, Neurokinin-1 metabolism, Receptors, Opioid, delta drug effects, Receptors, Opioid, delta metabolism, Receptors, Opioid, mu agonists, Receptors, Opioid, mu drug effects, Spinal Cord drug effects, Spinal Cord Injuries drug therapy, Analgesics pharmacology, Analgesics, Opioid pharmacology, Ligands
- Abstract
The lower efficacy of opioids in neuropathic pain may be due to the increased activity of pronociceptive systems such as substance P. We present evidence to support this hypothesis in this work from the spinal cord in a neuropathic pain model in mice. Biochemical analysis confirmed the elevated mRNA and protein level of pronociceptive substance P, the major endogenous ligand of the neurokinin-1 (NK1) receptor, in the lumbar spinal cord of chronic constriction injury (CCI)-mice. To improve opioid efficacy in neuropathic pain, novel compounds containing opioid agonist and neurokinin 1 (NK1) receptor antagonist pharmacophores were designed. Structure-activity studies were performed on opioid agonist/NK1 receptor antagonist hybrid peptides by modification of the C-terminal amide substituents. All compounds were evaluated for their affinity and in vitro activity at the mu opioid (MOP) and delta opioid (DOP) receptors, and for their affinity and antagonist activity at the NK1 receptor. On the basis of their in vitro profiles, the analgesic properties of two new bifunctional hybrids were evaluated in naive and CCI-mice, representing models for acute and neuropathic pain, respectively. The compounds were administered to the spinal cord by lumbar puncture. In naive mice, the single pharmacophore opioid parent compounds provided better analgesic results, as compared to the hybrids (max 70% MPE), raising the acute pain threshold close to 100% MPE. On the other hand, the opioid parents gave poor analgesic effects under neuropathic pain conditions, while the best hybrid delivered robust (close to 100% MPE) and long lasting alleviation of both tactile and thermal hypersensitivity. The results presented emphasize the potential of opioid/NK1 hybrids in view of analgesia under nerve injury conditions.
- Published
- 2017
- Full Text
- View/download PDF
13. Comparison of the Expression Changes after Botulinum Toxin Type A and Minocycline Administration in Lipopolysaccharide-Stimulated Rat Microglial and Astroglial Cultures.
- Author
-
Piotrowska A, Popiolek-Barczyk K, Pavone F, and Mika J
- Subjects
- Animals, Astrocytes metabolism, Botulinum Toxins, Type A therapeutic use, Cell Culture Techniques, Cell Survival drug effects, Cytokines drug effects, Cytokines metabolism, Interleukin 1 Receptor Antagonist Protein metabolism, Interleukin-1beta metabolism, Interleukin-6 metabolism, Interleukin-8 metabolism, MAP Kinase Signaling System drug effects, Matrix Metalloproteinase 9 drug effects, Microglia metabolism, Minocycline therapeutic use, Myeloid Differentiation Factor 88 metabolism, NF-kappa B metabolism, Nitric Oxide Synthase Type II metabolism, Rats, Signal Transduction drug effects, Synaptosomal-Associated Protein 25, Toll-Like Receptor 2 metabolism, Toll-Like Receptor 4 metabolism, Vesicular Transport Proteins metabolism, Astrocytes drug effects, Botulinum Toxins, Type A administration & dosage, Botulinum Toxins, Type A pharmacology, Lipopolysaccharides pharmacology, Microglia drug effects, Minocycline administration & dosage, Minocycline pharmacology
- Abstract
Botulinum neurotoxin type A (BoNT/A) and minocycline are potent drugs used in clinical therapies. The primary molecular mechanism of BoNT/A is the cleavage of SNARE proteins, which prevents cells from releasing neurotransmitters from vesicles, while the effects of minocycline are related to the inhibition of p38 activation. Both BoNT/A and minocycline exhibit analgesic effects, however, their direct impact on glial cells is not fully known. Therefore, the aim of the present study was to determine the effects of those drugs on microglial and astroglial activity after lipopolysaccharide (LPS) stimulation and their potential synergistic action. Our results show that BoNT/A and minocycline influenced primary microglial cells by inhibiting intracellular signaling pathways, such as p38, ERK1/2, NF-κB, and the release of pro-inflammatory factors, including IL-1β, IL-18, IL-6, and NOS2. We have revealed that, in contrast to minocycline, BoNT/A treatment did not decrease LPS-induced release of pro-inflammatory factors in the astroglia. In addition, BoNT/A decreased SNAP-23 in both types of glial cells and also SNAP-25 expressed only in astrocytes. Moreover, BoNT/A increased TLR2 and its adaptor protein MyD88, but not TLR4 exclusively in microglial cells. Furthermore, we have shown the impact of BoNT/A on microglial and astroglial cells, with a particular emphasis on its molecular target, TLR2. In contrast, minocycline did not affect any of those factors. We have revealed that despite of different molecular targets, minocycline, and BoNT/A reduced the release of microglia-derived pro-inflammatory factors. In conclusion, we have shown that BoNT/A and minocycline are effective drugs for the management of neuroinflammation by dampening the activation of microglial cells, with minocycline also affecting astroglial activity.
- Published
- 2017
- Full Text
- View/download PDF
14. Biphalin, a Dimeric Enkephalin, Alleviates LPS-Induced Activation in Rat Primary Microglial Cultures in Opioid Receptor-Dependent and Receptor-Independent Manners.
- Author
-
Popiolek-Barczyk K, Piotrowska A, Makuch W, and Mika J
- Subjects
- Animals, Cell Survival drug effects, Cells, Cultured, Inflammation chemically induced, Inflammation metabolism, Inflammation Mediators metabolism, Lipopolysaccharides, Male, Nitric Oxide metabolism, Nociception drug effects, Nociception physiology, Rats, Wistar, Enkephalins administration & dosage, Microglia drug effects, Microglia metabolism, Neuralgia metabolism, Receptors, Opioid metabolism
- Abstract
Neuropathic pain is relatively less responsive to opioids than other types of pain, which is possibly due to a disrupted opioid system partially caused by the profound microglial cell activation that underlines neuroinflammation. We demonstrated that intrathecally injected biphalin, a dimeric enkephalin analog, diminished symptoms of neuropathy in a preclinical model of neuropathic pain in rats (CCI, chronic constriction injury of the sciatic nerve) at day 12 postinjury. Using primary microglial cell cultures, we revealed that biphalin did not influence cell viability but diminished NO production and expression of Iba1 in LPS-stimulated cells. Biphalin also diminished MOP receptor level, as well as pronociceptive mediators (iNOS, IL-1 β , and IL-18) in an opioid receptor-dependent manner, and it was correlated with diminished p-NF- κ B, p-I κ B, p-p38MAPK, and TRIF levels. Biphalin reduced IL-6, IL-10, TNF α , p-STAT3, and p-ERK1/2 and upregulated SOCS3, TLR4, and MyD88; however, this effect was not reversed by naloxone pretreatment. Our study provides evidence that biphalin diminishes neuropathy symptoms, which might be partially related to reduced pronociceptive mediators released by activated microglia. Biphalin may be a putative drug for future pain therapy, especially for the treatment of neuropathic pain, when the lower analgesic effects of morphine are correlated with profound microglial cell activation.
- Published
- 2017
- Full Text
- View/download PDF
15. Anti-inflammatory properties of tianeptine on lipopolysaccharide-induced changes in microglial cells involve toll-like receptor-related pathways.
- Author
-
Slusarczyk J, Trojan E, Glombik K, Piotrowska A, Budziszewska B, Kubera M, Popiolek-Barczyk K, Lason W, Mika J, and Basta-Kaim A
- Subjects
- Animals, Cytokines metabolism, NF-kappa B metabolism, Nitric Oxide metabolism, Nitric Oxide Synthase Type II metabolism, Rats, Sprague-Dawley, Toll-Like Receptor 4 metabolism, Tumor Necrosis Factor-alpha metabolism, Anti-Inflammatory Agents pharmacology, Inflammation drug therapy, Lipopolysaccharides pharmacology, Microglia drug effects, Thiazepines pharmacology, Toll-Like Receptor 4 drug effects
- Abstract
Accumulating evidence suggests that activation of microglia plays a key role in the pathogenesis of depression. Activated microglia produce a wide range of factors whose prolonged or excessive release may lead to brain disorders. Thus, the inhibition of microglial cells may be beneficial in the treatment of depressive diseases. Tianeptine is an atypical antidepressant drug with proven clinical efficacy, but its mechanism of action remains still not fully understood. In the present study, using microglial cultures we investigated whether tianeptine modifies microglial activation after lipopolysaccharide (LPS) stimulation and which intracellular pathways are involved in the activity of this antidepressant. Our study shows that tianeptine attenuated the LPS-evoked inflammatory activation of microglia by decreasing the expression of proinflammatory cytokines such as IL-1β, IL-18, IL-6 and tumor necrosis factor α (TNF-α), the release of nitric oxide (NO) and reactive oxygen species (ROS) as well as the expression of inducible nitric oxide synthase. Analyses of signaling pathways demonstrate that tianeptine led to the suppression of LPS-induced TLR4 expression and ERK1/2 phosphorylation. Furthermore, our study reveals the inhibitory impact of tianeptine on caspase-3-induced PKCδ degradation and consequently on the activation of NF-κB factor in microglial cells. Taken together, present results show anti-inflammatory properties of tianeptine in microglial cultures stimulated by LPS. This study provides evidence that the inhibition of microglial activation may underlie the therapeutic activity of tianeptine. Our findings show the anti-inflammatory effect of tianeptine (TIA) in lipopolisaccharide (LPS)-stimulated microglial cells. The beneficial tianeptine action is mediated through the inhibition of Toll-like receptor 4 (TLR4) expression as well as the TLR4-related pathways: extracellular signal-regulated kinase 1/2 (ERK1/2), caspase-3-dependent protein kinase δ (PKCδ) cleavage and the expression of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). These findings may provide a new therapeutic strategy for treatment of disorders based on neuroinflammation, including depression., (© 2015 International Society for Neurochemistry.)
- Published
- 2016
- Full Text
- View/download PDF
16. Targeting the Microglial Signaling Pathways: New Insights in the Modulation of Neuropathic Pain.
- Author
-
Popiolek-Barczyk K and Mika J
- Subjects
- Central Nervous System metabolism, Humans, Mitogen-Activated Protein Kinases metabolism, NF-kappa B metabolism, Neuralgia metabolism, Neurons metabolism, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, STAT Transcription Factors metabolism, Signal Transduction, Microglia metabolism, Neuralgia pathology
- Abstract
The microglia, once thought only to be supporting cells of the central nervous system (CNS), are now recognized to play essential roles in many pathologies. Many studies within the last decades indicated that the neuro-immune interaction underlies the generation and maintenance of neuropathic pain. Through a large number of receptors and signaling pathways, the microglial cells communicate with neurons, astrocytes and other cells, including those of the immune system. A disturbance or loss of CNS homeostasis causes rapid responses of the microglia, which undergo a multistage activation process. The activated microglia change their cell shapes and gene expression profiles, which induce proliferation, migration, and the production of pro- or antinociceptive factors. The cells release a large number of mediators that can act in a manner detrimental or beneficial to the surrounding cells and can indirectly alter the nociceptive signals. This review discusses the most important microglial intracellular signaling cascades (MAPKs, NF-kB, JAK/STAT, PI3K/Akt) that are essential for neuropathic pain development and maintenance. Our objective was to identify new molecular targets that may result in the development of powerful tools to control the signaling associated with neuropathic pain.
- Published
- 2016
- Full Text
- View/download PDF
17. PD98059 Influences Immune Factors and Enhances Opioid Analgesia in Model of Neuropathy.
- Author
-
Rojewska E, Popiolek-Barczyk K, Kolosowska N, Piotrowska A, Zychowska M, Makuch W, Przewlocka B, and Mika J
- Subjects
- Analgesics, Opioid therapeutic use, Animals, Cytokines metabolism, Disease Models, Animal, Drug Synergism, Extracellular Signal-Regulated MAP Kinases metabolism, JNK Mitogen-Activated Protein Kinases metabolism, Male, NF-kappa B metabolism, Neuralgia physiopathology, Nociception drug effects, Rats, Rats, Wistar, p38 Mitogen-Activated Protein Kinases metabolism, Analgesics, Opioid pharmacology, Flavonoids pharmacology, Neuralgia drug therapy, Neuralgia immunology
- Abstract
Neuropathic pain treatment remains challenging due to ineffective therapy and resistance to opioid analgesia. Mitogen-activated protein kinase kinase (MAPKK) have been identified as the crucial regulators of pro- and antinociceptive factors. We used PD98059, an inhibitor of the MAPKK family members MEK1/2. The aim of study was to examine the influence of single and/or repeated PD98059 on nociception and opioid effectiveness in neuropathy. Moreover, we examined how PD98059 influences selected members of cellular pathways and cytokines. The PD98059 (2.5 mcg) was intrathecally preemptively administered before chronic constriction injury (CCI), and then once daily for 7 days. Additionally, at day 7 after CCI the PD98059-treated rats received a single injection of opioids. Using Western blot and qRT-PCR techniques in PD98059-treated rats we analyzed the mRNA and/or protein level of p38, ERK1/2, JNK, NF-kappaB, IL-1beta, IL-6, iNOS and IL-10 in the lumbar spinal cord. Our results indicate that PD98059 has an analgesic effects and potentiates morphine and/or buprenorphine analgesia. Parallel we observed that PD98059 inhibit upregulation of the CCI-elevated p38, ERK1/2, JNK and NF-kappaB protein levels. Moreover, PD98059 also prevented increase of pro- (IL-1beta, IL-6, and iNOS) but enhances anti-nociceptive (IL-10) factors. Summing up, PD98059 diminished pain and increased the effectiveness of opioids in neuropathy. The inhibition of MEKs might inactivate a variety of cell signaling pathways that are implicated in nociception.
- Published
- 2015
- Full Text
- View/download PDF
18. Anandamide, Acting via CB2 Receptors, Alleviates LPS-Induced Neuroinflammation in Rat Primary Microglial Cultures.
- Author
-
Malek N, Popiolek-Barczyk K, Mika J, Przewlocka B, and Starowicz K
- Subjects
- Animals, Arachidonic Acids therapeutic use, Cells, Cultured, Encephalitis chemically induced, Encephalitis drug therapy, Endocannabinoids therapeutic use, Immunologic Factors therapeutic use, Lipopolysaccharides, Nitric Oxide metabolism, Polyunsaturated Alkamides therapeutic use, Rats, Rats, Wistar, Receptors, Cannabinoid metabolism, Receptors, G-Protein-Coupled metabolism, Arachidonic Acids pharmacology, Encephalitis metabolism, Endocannabinoids pharmacology, Immunologic Factors pharmacology, Microglia drug effects, Microglia metabolism, Polyunsaturated Alkamides pharmacology, Receptor, Cannabinoid, CB2 metabolism
- Abstract
Microglial activation is a polarized process divided into potentially neuroprotective phenotype M2 and neurotoxic phenotype M1, predominant during chronic neuroinflammation. Endocannabinoid system provides an attractive target to control the balance between microglial phenotypes. Anandamide as an immune modulator in the central nervous system acts via not only cannabinoid receptors (CB1 and CB2) but also other targets (e.g., GPR18/GPR55). We studied the effect of anandamide on lipopolysaccharide-induced changes in rat primary microglial cultures. Microglial activation was assessed based on nitric oxide (NO) production. Analysis of mRNA was conducted for M1 and M2 phenotype markers possibly affected by the treatment. Our results showed that lipopolysaccharide-induced NO release in microglia was significantly attenuated, with concomitant downregulation of M1 phenotypic markers, after pretreatment with anandamide. This effect was not sensitive to CB1 or GPR18/GPR55 antagonism. Administration of CB2 antagonist partially abolished the effects of anandamide on microglia. Interestingly, administration of a GPR18/GPR55 antagonist by itself suppressed NO release. In summary, we showed that the endocannabinoid system plays a crucial role in the management of neuroinflammation by dampening the activation of an M1 phenotype. This effect was primarily controlled by the CB2 receptor, although functional cross talk with GPR18/GPR55 may occur.
- Published
- 2015
- Full Text
- View/download PDF
19. Parthenolide Relieves Pain and Promotes M2 Microglia/Macrophage Polarization in Rat Model of Neuropathy.
- Author
-
Popiolek-Barczyk K, Kolosowska N, Piotrowska A, Makuch W, Rojewska E, Jurga AM, Pilat D, and Mika J
- Subjects
- Analgesics therapeutic use, Animals, Cell Polarity drug effects, Cells, Cultured, Chronic Disease, Hyperalgesia drug therapy, Hyperalgesia metabolism, Macrophages metabolism, Male, Microglia metabolism, Neuralgia drug therapy, Rats, Rats, Wistar, Sciatic Neuropathy drug therapy, Sesquiterpenes therapeutic use, Signal Transduction drug effects, Analgesics administration & dosage, Macrophages drug effects, Microglia drug effects, Neuralgia metabolism, Sciatic Neuropathy metabolism, Sesquiterpenes administration & dosage
- Abstract
Neuropathic pain treatment remains a challenge because pathomechanism is not fully understood. It is believed that glial activation and increased spinal nociceptive factors are crucial for neuropathy. We investigated the effect of parthenolide (PTL) on the chronic constriction injury to the sciatic nerve (CCI)-induced neuropathy in rat. We analyzed spinal changes in glial markers and M1 and M2 polarization factors, as well as intracellular signaling pathways. PTL (5 µg; i.t.) was preemptively and then daily administered for 7 days after CCI. PTL attenuated the allodynia and hyperalgesia and increased the protein level of IBA1 (a microglial/macrophage marker) but did not change GFAP (an astrocyte marker) on day 7 after CCI. PTL reduced the protein level of M1 (IL-1β, IL-18, and iNOS) and enhanced M2 (IL-10, TIMP1) factors. In addition, it downregulated the phosphorylated form of NF-κB, p38MAPK, and ERK1/2 protein level and upregulated STAT3. In primary microglial cell culture we have shown that IL-1β, IL-18, iNOS, IL-6, IL-10, and TIMP1 are of microglial origin. Summing up, PTL directly or indirectly attenuates neuropathy symptoms and promotes M2 microglia/macrophages polarization. We suggest that neuropathic pain therapies should be shifted from blanketed microglia/macrophage suppression toward maintenance of the balance between neuroprotective and neurotoxic microglia/macrophage phenotypes.
- Published
- 2015
- Full Text
- View/download PDF
20. Involvement of pro- and antinociceptive factors in minocycline analgesia in rat neuropathic pain model.
- Author
-
Rojewska E, Popiolek-Barczyk K, Jurga AM, Makuch W, Przewlocka B, and Mika J
- Subjects
- Animals, Animals, Newborn, Anti-Inflammatory Agents, Non-Steroidal therapeutic use, Cells, Cultured, Cytokines genetics, Disease Models, Animal, Ganglia, Spinal pathology, Gene Expression Regulation drug effects, Hyperalgesia drug therapy, Hyperalgesia etiology, Male, Matrix Metalloproteinase 2 genetics, Matrix Metalloproteinase 9 genetics, Microglia drug effects, Neurons drug effects, Neurons metabolism, Pain Measurement, Pain Threshold drug effects, Rats, Rats, Wistar, Sciatica pathology, Spinal Cord pathology, Tissue Inhibitor of Metalloproteinase-1 genetics, Analgesics therapeutic use, Cytokines metabolism, Matrix Metalloproteinase 2 metabolism, Matrix Metalloproteinase 9 metabolism, Minocycline therapeutic use, Sciatica drug therapy, Tissue Inhibitor of Metalloproteinase-1 metabolism
- Abstract
In neuropathic pain the repeated minocycline treatment inhibited the mRNA and protein expression of the microglial markers and metalloproteinase-9 (MMP-9). The minocycline diminished the pronociceptive (IL-6, IL-18), but not antinociceptive (IL-1alpha, IL-4, IL-10) cytokines at the spinal cord level. In vitro primary cell culture studies have shown that MMP-9, TIMP-1, IL-1beta, IL-1alpha, IL-6, IL-10, and IL-18 are of microglial origin. Minocycline reduces the production of pronociceptive factors, resulting in a more potent antinociceptive effect. This change in the ratio between pro- and antinociceptive factors, in favour of the latter may be the mechanism of minocycline analgesia in neuropathy., (Copyright © 2014. Published by Elsevier B.V.)
- Published
- 2014
- Full Text
- View/download PDF
21. Inhibition of intracellular signaling pathways NF-κB and MEK1/2 attenuates neuropathic pain development and enhances morphine analgesia.
- Author
-
Popiolek-Barczyk K, Makuch W, Rojewska E, Pilat D, and Mika J
- Subjects
- Analgesics, Opioid pharmacology, Animals, Disease Models, Animal, Drug Synergism, Hyperalgesia drug therapy, MAP Kinase Kinase 1 antagonists & inhibitors, MAP Kinase Kinase 1 metabolism, MAP Kinase Kinase 2 antagonists & inhibitors, MAP Kinase Kinase 2 metabolism, Male, Morphine pharmacology, NF-kappa B antagonists & inhibitors, NF-kappa B metabolism, Neuralgia physiopathology, Rats, Rats, Wistar, Reverse Transcriptase Polymerase Chain Reaction, Sciatic Nerve injuries, Signal Transduction drug effects, Analgesics pharmacology, Butadienes pharmacology, Neuralgia drug therapy, Nitriles pharmacology, Sesquiterpenes pharmacology
- Abstract
Background: Neuropathic pain is clinically challenging because it is resistant to alleviation by morphine. The nuclear factor κB (NF-κB) and mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathways may be involved in the development of neuropathic pain. The aim of our study was to examine the influence of a chronic, intrathecal administration of parthenolide (PTL, inhibitor of NF-κB) and U0126 (inhibitor of MEK1/2) on nociception and morphine effectiveness in a rat model of neuropathy., Methods: The chronic constriction injury of the sciatic nerve in Wistar rats was performed. PTL and U0126 were injected chronic intrathecally and morphine was injected once at day 7. To evaluate allodynia and hyperalgesia, the von Frey and cold plate tests were used, respectively. The experiments were carried out according to IASP rules. Using qRT-PCR we analyzed mRNAs of μ-(mor), δ-(dor) and κ-(kor)-opioid receptors in the lumbar spinal cord after drugs administration., Results: The administration of PTL and U0126 decreased allodynia and hyperalgesia and significantly potentiated morphine effect. The mor, dor and kor mRNAs were down-regulated 7 days after injury in the ipsilateral spinal cord. The PTL and U0126 significantly up-regulated the mRNA levels of all opioid receptors. The levels of mor and dor mRNAs were much higher compared to those in naïve, but only the kor levels returned to control values., Conclusions: These results indicate that the inhibition of the NF-κB pathway has better analgesic effects. Both inhibitors similarly potentiate morphine analgesia, which parallels the up-regulation of both mor and dor mRNAs expression spinal levels of the model of neuropathy., (Copyright © 2014 Institute of Pharmacology, Polish Academy of Sciences. Published by Elsevier Urban & Partner Sp. z o.o. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
22. Delta-opioid receptor analgesia is independent of microglial activation in a rat model of neuropathic pain.
- Author
-
Mika J, Popiolek-Barczyk K, Rojewska E, Makuch W, Starowicz K, and Przewlocka B
- Subjects
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer administration & dosage, 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer therapeutic use, Analgesics, Opioid administration & dosage, Animals, Anti-Bacterial Agents administration & dosage, Anti-Bacterial Agents therapeutic use, Cells, Cultured, Enkephalin, Ala(2)-MePhe(4)-Gly(5)- administration & dosage, Enkephalin, Ala(2)-MePhe(4)-Gly(5)- therapeutic use, Gene Expression Regulation drug effects, Male, Microglia cytology, Microglia metabolism, Minocycline administration & dosage, Minocycline therapeutic use, Morphine administration & dosage, Morphine therapeutic use, Rats, Wistar, Receptors, Opioid, delta genetics, Analgesics, Opioid therapeutic use, Microglia drug effects, Neuralgia drug therapy, Receptors, Opioid, delta agonists, Receptors, Opioid, delta metabolism
- Abstract
The analgesic effect of delta-opioid receptor (DOR) ligands in neuropathic pain is not diminished in contrast to other opioid receptor ligands, which lose their effectiveness as analgesics. In this study, we examine whether this effect is related to nerve injury-induced microglial activation. We therefore investigated the influence of minocycline-induced inhibition of microglial activation on the analgesic effects of opioid receptor agonists: morphine, DAMGO, U50,488H, DPDPE, Deltorphin II and SNC80 after chronic constriction injury (CCI) to the sciatic nerve in rats. Pre-emptive and repeated administration of minocycline (30 mg/kg, i.p.) over 7 days significantly reduced allodynia and hyperalgesia as measured on day 7 after CCI. The antiallodynic and antihyperalgesic effects of intrathecally (i.t.) administered morphine (10-20 µg), DAMGO (1-2 µg) and U50,488H (25-50 µg) were significantly potentiated in rats after minocycline, but no such changes were observed after DPDPE (10-20 µg), deltorphin II (1.5-15 µg) and SNC80 (10-20 µg) administration. Additionally, nerve injury-induced down-regulation of all types of opioid receptors in the spinal cord and dorsal root ganglia was not influenced by minocycline, which indicates that the effects of opioid ligands are dependent on other changes, presumably neuroimmune interactions. Our study of rat primary microglial cell culture using qRT-PCR, Western blotting and immunocytochemistry confirmed the presence of mu-opioid receptors (MOR) and kappa-opioid receptors (KOR), further we provide the first evidence for the lack of DOR on microglial cells. In summary, DOR analgesia is different from analgesia induced by MOR and KOR receptors because it does not dependent on injury-induced microglial activation. DOR agonists appear to be the best candidates for new drugs to treat neuropathic pain.
- Published
- 2014
- Full Text
- View/download PDF
23. Minocycline enhances the effectiveness of nociceptin/orphanin FQ during neuropathic pain.
- Author
-
Popiolek-Barczyk K, Rojewska E, Jurga AM, Makuch W, Zador F, Borsodi A, Piotrowska A, Przewlocka B, and Mika J
- Subjects
- Animals, Cells, Cultured, Gene Expression Regulation drug effects, Male, Microglia drug effects, Microglia pathology, Minocycline administration & dosage, Minocycline pharmacology, Models, Biological, Neuralgia pathology, RNA, Messenger genetics, RNA, Messenger metabolism, Rats, Wistar, Receptors, Opioid genetics, Receptors, Opioid metabolism, Signal Transduction drug effects, Signal Transduction genetics, Spinal Cord drug effects, Spinal Cord pathology, Treatment Outcome, Nociceptin Receptor, Nociceptin, Minocycline therapeutic use, Neuralgia drug therapy, Opioid Peptides therapeutic use
- Abstract
Nociceptin/orphanin FQ (N/OFQ) antinociception, which is mediated selectively by the N/OFQ peptide receptor (NOP), was demonstrated in pain models. In this study, we determine the role of activated microglia on the analgesic effects of N/OFQ in a rat model of neuropathic pain induced by chronic constriction injury (CCI) to the sciatic nerve. Repeated 7-day administration of minocycline (30 mg/kg i.p.), a drug that affects microglial activation, significantly reduced pain in CCI-exposed rats and it potentiates the analgesic effects of administered N/OFQ (2.5-5 μg i.t.). Minocycline also downregulates the nerve injury-induced upregulation of NOP protein in the dorsal lumbar spinal cord. Our in vitro study showed that minocycline reduced NOP mRNA, but not protein, level in rat primary microglial cell cultures. In [(35)S]GTPγS binding assays we have shown that minocycline increases the spinal N/OFQ-stimulated NOP signaling. We suggest that the modulation of the N/OFQ system by minocycline is due to the potentiation of its neuronal antinociceptive activity and weakening of the microglial cell activation. This effect is beneficial for pain relief, and these results suggest new targets for the development of drugs that are effective against neuropathic pain.
- Published
- 2014
- Full Text
- View/download PDF
24. Importance of glial activation in neuropathic pain.
- Author
-
Mika J, Zychowska M, Popiolek-Barczyk K, Rojewska E, and Przewlocka B
- Subjects
- Animals, Central Nervous System drug effects, Central Nervous System pathology, Humans, Morphine pharmacology, Morphine therapeutic use, Neuralgia drug therapy, Neuralgia immunology, Neuroglia drug effects, Neuralgia pathology, Neuroglia pathology
- Abstract
Glia plays a crucial role in the maintenance of neuronal homeostasis in the central nervous system. The microglial production of immune factors is believed to play an important role in nociceptive transmission. Pain may now be considered a neuro-immune disorder, since it is known that the activation of immune and immune-like glial cells in the dorsal root ganglia and spinal cord results in the release of both pro- and anti-inflammatory cytokines, as well as algesic and analgesic mediators. In this review we presented an important role of cytokines (IL-1alfa, IL-1beta, IL-2, IL-4, IL-6, IL-10, IL-15, IL-18, TNFalpha, IFNgamma, TGF-beta 1, fractalkine and CCL2); complement components (C1q, C3, C5); metaloproteinases (MMP-2,-9) and many other factors, which become activated on spinal cord and DRG level under neuropathic pain. We discussed the role of the immune system in modulating chronic pain. At present, unsatisfactory treatment of neuropathic pain will seek alternative targets for new drugs and it is possible that anti-inflammatory factors like IL-10, IL-4, IL-1alpha, TGF-beta 1 would fulfill this role. Another novel approach for controlling neuropathic pain can be pharmacological attenuation of glial and immune cell activation. It has been found that propentofylline, pentoxifylline, minocycline and fluorocitrate suppress the development of neuropathic pain. The other way of pain control can be the decrease of pro-nociceptive agents like transcription factor synthesis (NF-kappaB, AP-1); kinase synthesis (MEK, p38MAPK, JNK) and protease activation (cathepsin S, MMP9, MMP2). Additionally, since it is known that the opioid-induced glial activation opposes opioid analgesia, some glial inhibitors, which are safe and clinically well tolerated, are proposed as potential useful ko-analgesic agents for opioid treatment of neuropathic pain. This review pointed to some important mechanisms underlying the development of neuropathic pain, which led to identify some possible new approaches to the treatment of neuropathic pain, based on the more comprehensive knowledge of the interaction between the nervous system and glial and immune cells., (© 2013 Elsevier B.V. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.