Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Biol Open ; 12(6)2023 06 15.
Article in English | MEDLINE | ID: mdl-37259940

ABSTRACT

Early phase diabetes is often accompanied by pain sensitization. In Drosophila, the insulin receptor (InR) regulates the persistence of injury-induced thermal nociceptive sensitization. Whether Drosophila InR also regulates the persistence of mechanical nociceptive sensitization remains unclear. Mice with a sensory neuron deletion of the insulin receptor (Insr) show normal nociceptive baselines; however, it is uncertain whether deletion of Insr in nociceptive sensory neurons leads to persistent nociceptive hypersensitivity. In this study, we used fly and mouse nociceptive sensitization models to address these questions. In flies, InR mutants and larvae with sensory neuron-specific expression of RNAi transgenes targeting InR exhibited persistent mechanical hypersensitivity. Mice with a specific deletion of the Insr gene in Nav1.8+ nociceptive sensory neurons showed nociceptive thermal and mechanical baselines similar to controls. In an inflammatory paradigm, however, these mutant mice showed persistent mechanical (but not thermal) hypersensitivity, particularly in female mice. Mice with the Nav1.8+ sensory neuron-specific deletion of Insr did not show metabolic abnormalities typical of a defect in systemic insulin signaling. Our results show that some aspects of the regulation of nociceptive hypersensitivity by the insulin receptor are shared between flies and mice and that this regulation is likely independent of metabolic effects.


Subject(s)
Drosophila Proteins , Receptor, Insulin , Animals , Mice , Female , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Nociception/physiology , Drosophila/metabolism , Sensory Receptor Cells/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism
2.
Pain ; 161(10): 2344-2352, 2020 10.
Article in English | MEDLINE | ID: mdl-32427749

ABSTRACT

Understanding the mechanisms that drive transition from acute to chronic pain is essential to identify new therapeutic targets. The importance of endogenous resolution pathways acting as a "brake" to prevent development of chronic pain has been largely ignored. We examined the role of interleukin-10 (IL-10) in resolution of neuropathic pain induced by cisplatin. In search of an underlying mechanism, we studied the effect of cisplatin and IL-10 on spontaneous activity (SA) in dorsal root ganglia neurons. Cisplatin (2 mg/kg daily for 3 days) induced mechanical hypersensitivity that resolved within 3 weeks. In both sexes, resolution of mechanical hypersensitivity was delayed in Il10 mice, in WT mice treated intrathecally with neutralizing anti-IL-10 antibody, and in mice with cell-targeted deletion of IL-10R1 on advillin-positive sensory neurons. Electrophysiologically, small- to medium-sized dorsal root ganglia neurons from cisplatin-treated mice displayed an increase in the incidence of SA. Cisplatin treatment also depolarized the resting membrane potential, and decreased action potential voltage threshold and rheobase, while increasing ongoing activity at -45 mV and the amplitude of depolarizing spontaneous fluctuations. In vitro addition of IL-10 (10 ng/mL) reversed the effect of cisplatin on SA and on the depolarizing spontaneous fluctuation amplitudes, but unexpectedly had little effect on the other electrophysiological parameters affected by cisplatin. Collectively, our findings challenge the prevailing concept that IL-10 resolves pain solely by dampening neuroinflammation and demonstrate in a model of chemotherapy-induced neuropathic pain that endogenous IL-10 prevents transition to chronic pain by binding to IL-10 receptors on sensory neurons to regulate their activity.


Subject(s)
Hyperalgesia/metabolism , Action Potentials , Animals , Cisplatin/toxicity , Female , Ganglia, Spinal , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Interleukin-10 , Male , Mice , Sensory Receptor Cells
3.
Proc Natl Acad Sci U S A ; 117(10): 5463-5471, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32079726

ABSTRACT

Chronic pain is a major clinical problem of which the mechanisms are incompletely understood. Here, we describe the concept that PI16, a protein of unknown function mainly produced by fibroblasts, controls neuropathic pain. The spared nerve injury (SNI) model of neuropathic pain increases PI16 protein levels in fibroblasts in dorsal root ganglia (DRG) meninges and in the epi/perineurium of the sciatic nerve. We did not detect PI16 expression in neurons or glia in spinal cord, DRG, and nerve. Mice deficient in PI16 are protected against neuropathic pain. In vitro, PI16 promotes transendothelial leukocyte migration. In vivo, Pi16-/- mice show reduced endothelial barrier permeability, lower leukocyte infiltration and reduced activation of the endothelial barrier regulator MLCK, and reduced phosphorylation of its substrate MLC2 in response to SNI. In summary, our findings support a model in which PI16 promotes neuropathic pain by mediating a cross-talk between fibroblasts and the endothelial barrier leading to barrier opening, cellular influx, and increased pain. Its key role in neuropathic pain and its limited cellular and tissue distribution makes PI16 an attractive target for pain management.


Subject(s)
Fibroblasts/enzymology , Neuralgia/genetics , Proteinase Inhibitory Proteins, Secretory/genetics , Animals , Cell Movement , Chronic Pain , Disease Models, Animal , Endothelial Cells/physiology , Ganglia, Spinal , Leukocytes/physiology , Meninges/cytology , Mice, Knockout , Peripheral Nerve Injuries/physiopathology , Sciatic Nerve/enzymology
4.
Brain Behav Immun ; 79: 319-325, 2019 07.
Article in English | MEDLINE | ID: mdl-30953771

ABSTRACT

Cancer and its treatment are associated with neurotoxic side effects, including cognitive dysfunction, altered functional connectivity in the brain and structural abnormalities in white matter. There is evidence that cancer and its treatment can accelerate aging. Tau is a microtubule associated protein that contributes to microtubule stability thereby playing a key role in neuronal function. Clustering of tau is commonly observed in the aged brain and is related to cognitive decline. We hypothesized that chemotherapy-induced cognitive impairment is associated with accelerated development of tau clustering in the brain as a sign of accelerated aging. We show for the first time that treatment of adult (7-8 month-old) male C57BL/6 mice with cisplatin results in reduced cognitive function and a marked increase in the number of large endogenous tau clusters in the hippocampus when assessed 4 months later. In contrast, we detected only few small tau clusters in the hippocampus of age-matched 11-12 month-old control mice. Astrocyte GFAP expression was increased in close vicinity to the tau clusters in cisplatin-treated mice. We did not detect changes in the microglial marker Iba-1 in the brain of mice treated with cisplatin. The accelerated formation of Tau-1 clusters in cisplatin-treated mice was associated with a decrease in the levels of the post-synaptic marker PSD95 and of the presynaptic marker synaptophysin in the hippocampus. We demonstrate here for the first time that chemotherapy markedly accelerates development of signs of tauopathy and loss of synaptic integrity in the hippocampus. These findings provide a mechanistic link between chemotherapy cognitive decline and accelerated aging in cancer survivors.


Subject(s)
Cisplatin/adverse effects , Cognitive Dysfunction/metabolism , Tauopathies/metabolism , Age Factors , Aging/metabolism , Animals , Brain/metabolism , Cognition/drug effects , Cognitive Dysfunction/etiology , Disease Models, Animal , Drug Therapy , Drug-Related Side Effects and Adverse Reactions , Glial Fibrillary Acidic Protein/analysis , Glial Fibrillary Acidic Protein/drug effects , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Tauopathies/etiology , tau Proteins/metabolism
5.
Sci Rep ; 8(1): 17400, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30479361

ABSTRACT

Patients treated for cancer frequently experience chemobrain, characterized by impaired memory and reduced attention. These deficits often persist after treatment, and no preventive or curative interventions exist. In mice, we assessed the effect of cisplatin chemotherapy on attention using the 5-choice serial reaction time task and on synaptic integrity. We also assessed the capacity of mesenchymal stem cells to normalize the characteristics of chemobrain. Mice were trained in the 5-choice serial reaction time task. After reaching advancement criteria at a 4-second stimulus time, they were treated with cisplatin followed by nasal administration of mesenchymal stem cells. Cisplatin reduced the percentage of correct responses due to an increase in omissions, indicating attention deficits. Mesenchymal stem cell treatment reversed these cisplatin-induced deficits in attention. Cisplatin also induced abnormalities in markers of synaptic integrity in the prefrontal cortex. Specifically, cisplatin decreased expression of the global presynaptic marker synaptophysin and the glutamatergic presynaptic marker vGlut2. Expression of the presynaptic GABAergic marker vGAT increased. Nasal mesenchymal stem cell administration normalized these markers of synaptic integrity. In conclusion, cisplatin induces long-lasting attention deficits that are associated with decreased synaptic integrity in the prefrontal cortex. Nasal administration of mesenchymal stem cells reversed these behavioural and structural deficits.


Subject(s)
Antineoplastic Agents/pharmacology , Attention Deficit Disorder with Hyperactivity/etiology , Cisplatin/pharmacology , Prefrontal Cortex/drug effects , Synaptic Transmission , Animals , Antineoplastic Agents/adverse effects , Attention Deficit Disorder with Hyperactivity/therapy , Cells, Cultured , Cisplatin/adverse effects , Male , Mesenchymal Stem Cell Transplantation/methods , Mice , Mice, Inbred C57BL , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiology , Synaptotagmins/genetics , Synaptotagmins/metabolism , Vesicular Glutamate Transport Protein 2/metabolism
6.
Acta Neuropathol Commun ; 6(1): 103, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30270813

ABSTRACT

Chemotherapy-induced cognitive impairment (CICI) is a commonly reported neurotoxic side effect of chemotherapy, occurring in up to 75% cancer patients. CICI manifests as decrements in working memory, executive functioning, attention, and processing speed, and greatly interferes with patients' daily performance and quality of life. Currently no treatment for CICI has been approved by the US Food and Drug Administration. We show here that treatment with a brain-penetrating histone deacetylase 6 (HDAC6) inhibitor for two weeks was sufficient to fully reverse cisplatin-induced cognitive impairments in male mice, as demonstrated in the Y-maze test of spontaneous alternation, the novel object/place recognition test, and the puzzle box test. Normalization of cognitive impairment was associated with reversal of cisplatin-induced synaptosomal mitochondrial deficits and restoration of synaptic integrity. Mechanistically, cisplatin induced deacetylation of the microtubule protein α-tubulin and hyperphosphorylation of the microtubule-associated protein tau. These cisplatin-induced changes were reversed by HDAC6 inhibition. Our data suggest that inhibition of HDAC6 restores microtubule stability and reverses tau phosphorylation, leading to normalization of synaptosomal mitochondrial function and synaptic integrity and thereby to reversal of CICI. Remarkably, our results indicate that short-term daily treatment with the HDAC6 inhibitor was sufficient to achieve prolonged reversal of established behavioral, structural and functional deficits induced by cisplatin. Because the beneficial effects of HDAC6 inhibitors as add-ons to cancer treatment have been demonstrated in clinical trials, selective targeting of HDAC6 with brain-penetrating inhibitors appears a promising therapeutic approach for reversing chemotherapy-induced neurotoxicity while enhancing tumor control.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Cognitive Dysfunction , Enzyme Inhibitors/therapeutic use , Histone Deacetylase 6/metabolism , Tauopathies/enzymology , Animals , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/enzymology , Disease Models, Animal , Disks Large Homolog 4 Protein/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/blood , Green Fluorescent Proteins/metabolism , Histone Deacetylase 6/ultrastructure , Hydroxamic Acids/blood , Hydroxamic Acids/therapeutic use , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Pyrimidines/blood , Pyrimidines/therapeutic use , Recombinant Fusion Proteins/metabolism , Synaptosomes/drug effects , Synaptosomes/metabolism , Synaptosomes/pathology , Synaptosomes/ultrastructure , Tauopathies/chemically induced , Tauopathies/drug therapy , Time Factors , Tubulin/metabolism , tau Proteins/metabolism
7.
Pain ; 159(5): 884-893, 2018 May.
Article in English | MEDLINE | ID: mdl-29369966

ABSTRACT

Chemotherapy-induced peripheral neuropathy (CIPN) is a major side effect of cancer treatment that significantly compromises quality of life of cancer patients and survivors. Identification of targets for pharmacological intervention to prevent or reverse CIPN is needed. We investigated exchange protein regulated by cAMP (Epac) as a potential target. Epacs are cAMP-binding proteins known to play a pivotal role in mechanical allodynia induced by nerve injury and inflammation. We demonstrate that global Epac1-knockout (Epac1-/-) male and female mice are protected against paclitaxel-induced mechanical allodynia. In addition, spinal cord astrocyte activation and intraepidermal nerve fiber (IENF) loss are significantly reduced in Epac1-/- mice as compared to wild-type mice. Moreover, Epac1-/- mice do not develop the paclitaxel-induced deficits in mitochondrial bioenergetics in the sciatic nerve that are a hallmark of CIPN. Notably, mice with cell-specific deletion of Epac1 in Nav1.8-positive neurons (N-Epac1-/-) also show reduced paclitaxel-induced mechanical allodynia, astrocyte activation, and IENF loss, indicating that CIPN develops downstream of Epac1 activation in nociceptors. The Epac-inhibitor ESI-09 reversed established paclitaxel-induced mechanical allodynia in wild-type mice even when dosing started 10 days after completion of paclitaxel treatment. In addition, oral administration of ESI-09 suppressed spinal cord astrocyte activation in the spinal cord and protected against IENF loss. Ex vivo, ESI-09 blocked paclitaxel-induced abnormal spontaneous discharges in dorsal root ganglion neurons. Collectively, these findings implicate Epac1 in nociceptors as a novel target for treatment of CIPN. This is clinically relevant because ESI-09 has the potential to reverse a debilitating and long-lasting side effect of cancer treatment.


Subject(s)
Antineoplastic Agents/adverse effects , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Hyperalgesia/drug therapy , Paclitaxel/adverse effects , Peripheral Nervous System Diseases/drug therapy , Animals , Astrocytes/drug effects , Disease Models, Animal , Female , Ganglia, Spinal/drug effects , Guanine Nucleotide Exchange Factors/genetics , Hydrazones/pharmacology , Hydrazones/therapeutic use , Hyperalgesia/etiology , Isoxazoles/pharmacology , Isoxazoles/therapeutic use , Male , Mice , Mice, Knockout , Nerve Fibers/drug effects , Pain Threshold/drug effects , Peripheral Nervous System Diseases/chemically induced , Spinal Cord/drug effects
8.
Brain Behav Immun ; 66: 94-102, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28709913

ABSTRACT

Pain and depression often co-occur, but the underlying mechanisms have not been elucidated. Here, we used the spared nerve injury (SNI) model in mice to induce both neuropathic pain and depression-like behavior. We investigated whether brain interleukin (IL)-1 signaling and activity of kynurenine 3-monoxygenase (KMO), a key enzyme for metabolism of kynurenine into the neurotoxic NMDA receptor agonist quinolinic acid, are necessary for comorbid neuropathic pain and depression-like behavior. SNI mice showed increased expression levels of Il1b and Kmo mRNA in the contralateral side of the brain. The SNI-induced increase of Kmo mRNA was associated with increased KMO protein and elevated quinolinic acid and reduced kynurenic acid in the contralateral hippocampus. The increase in KMO-protein in response to SNI mostly took place in hippocampal NeuN-positive neurons rather than microglia. Inhibition of brain IL-1 signaling by intracerebroventricular administration of IL-1 receptor antagonist after SNI prevented the increase in Kmo mRNA and depression-like behavior measured by forced swim test. However, inhibition of brain IL-1 signaling has no effect on mechanical allodynia. In addition, intracerebroventricular administration of the KMO inhibitor Ro 61-8048 abrogated depression-like behavior without affecting mechanical allodynia after SNI. We show for the first time that the development of depression-like behavior in the SNI model requires brain IL-1 signaling and activation of neuronal KMO, while pain is independent of this pathway. Inhibition of KMO may represent a promising target for treating depression.


Subject(s)
Depression/enzymology , Kynurenine 3-Monooxygenase/metabolism , Neuralgia/enzymology , Neurons/enzymology , Animals , Depression/complications , Disease Models, Animal , Hippocampus/enzymology , Hyperalgesia/complications , Hyperalgesia/enzymology , Interleukin-1/metabolism , Kynurenine 3-Monooxygenase/genetics , Male , Mice, Inbred C57BL , Microglia/enzymology , Neuralgia/complications , Peripheral Nerve Injuries/complications , Peripheral Nerve Injuries/enzymology , RNA, Messenger/metabolism , Signal Transduction , Up-Regulation
9.
Proc Natl Acad Sci U S A ; 113(11): 3036-41, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26929333

ABSTRACT

cAMP signaling plays a key role in regulating pain sensitivity. Here, we uncover a previously unidentified molecular mechanism in which direct phosphorylation of the exchange protein directly activated by cAMP 1 (EPAC1) by G protein kinase 2 (GRK2) suppresses Epac1-to-Rap1 signaling, thereby inhibiting persistent inflammatory pain. Epac1(-/-) mice are protected against inflammatory hyperalgesia in the complete Freund's adjuvant (CFA) model. Moreover, the Epac-specific inhibitor ESI-09 inhibits established CFA-induced mechanical hyperalgesia without affecting normal mechanical sensitivity. At the mechanistic level, CFA increased activity of the Epac target Rap1 in dorsal root ganglia of WT, but not of Epac1(-/-), mice. Using sensory neuron-specific overexpression of GRK2 or its kinase-dead mutant in vivo, we demonstrate that GRK2 inhibits CFA-induced hyperalgesia in a kinase activity-dependent manner. In vitro, GRK2 inhibits Epac1-to-Rap1 signaling by phosphorylation of Epac1 at Ser-108 in the Disheveled/Egl-10/pleckstrin domain. This phosphorylation event inhibits agonist-induced translocation of Epac1 to the plasma membrane, thereby reducing Rap1 activation. Finally, we show that GRK2 inhibits Epac1-mediated sensitization of the mechanosensor Piezo2 and that Piezo2 contributes to inflammatory mechanical hyperalgesia. Collectively, these findings identify a key role of Epac1 in chronic inflammatory pain and a molecular mechanism for controlling Epac1 activity and chronic pain through phosphorylation of Epac1 at Ser-108. Importantly, using the Epac inhibitor ESI-09, we validate Epac1 as a potential therapeutic target for chronic pain.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/physiology , Guanine Nucleotide Exchange Factors/physiology , Hyperalgesia/physiopathology , Inflammation/complications , Nociception/physiology , Pain/physiopathology , Amino Acid Sequence , Animals , Chronic Disease , Freund's Adjuvant/toxicity , Ganglia, Spinal/physiopathology , Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/genetics , Hyperalgesia/etiology , Inflammation/chemically induced , Ion Channels/physiology , Mechanoreceptors/physiology , Mice , Mice, Knockout , Molecular Sequence Data , Nerve Tissue Proteins/physiology , Pain/etiology , Pain Threshold/physiology , Phosphorylation , Phosphoserine/metabolism , Protein Interaction Mapping , Protein Processing, Post-Translational , Protein Structure, Tertiary , Signal Transduction , rap1 GTP-Binding Proteins/physiology
10.
Pain ; 156(11): 2184-2192, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26473292

ABSTRACT

Chemotherapy-induced peripheral neuropathy (CIPN) is a common side effect of cancer treatment. It is the most frequent cause of dose reduction or treatment discontinuation in patients treated for cancer with commonly used drugs including taxanes and platinum-based compounds. No FDA-approved treatments for CIPN are available. In rodents, CIPN is represented by peripheral mechanical allodynia in association with retraction of intraepidermal nerve fibers. The mechanism of chemotherapy-induced neurotoxicity is unclear, but it has been established that mitochondrial dysfunction is an important component of the dysregulation in peripheral sensory neurons. We have shown earlier that inhibition of mitochondrial p53 accumulation with the small compound pifithrin-µ (PFT-µ) prevents cerebral neuronal death in a rodent model of hypoxic-ischemic brain damage. We now explore whether PFT-µ is capable of preventing neuronal mitochondrial damage and CIPN in mice. We demonstrate for the first time that PFT-µ prevents both paclitaxel- and cisplatin-induced mechanical allodynia. Electron microscopic analysis of peripheral sensory nerves revealed that PFT-µ secured mitochondrial integrity in paclitaxel-treated mice. In addition, PFT-µ administration protects against chemotherapy-induced loss of intraepidermal nerve fibers in the paw. To determine whether neuroprotective treatment with PFT-µ would interfere with the antitumor effects of chemotherapy, ovarian tumor cells were cultured in vitro with PFT-µ and paclitaxel. Pifithrin-µ does not inhibit tumor cell death but even enhances paclitaxel-induced tumor cell death. These data are the first to identify PFT-µ as a potential therapeutic strategy for prevention of CIPN to combat one of the most devastating side effects of chemotherapy.


Subject(s)
Analgesics/therapeutic use , Antineoplastic Agents, Phytogenic/adverse effects , Paclitaxel/adverse effects , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/prevention & control , Sulfonamides/therapeutic use , Animals , Cisplatin/toxicity , Disease Models, Animal , Female , Ganglia, Spinal/pathology , Humans , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Mitochondria/pathology , Mitochondria/ultrastructure , Pain Measurement , Pain Threshold/drug effects , Peripheral Nervous System Diseases/pathology , Ubiquitin Thiolesterase/metabolism , Xenograft Model Antitumor Assays
11.
PLoS One ; 9(6): e100701, 2014.
Article in English | MEDLINE | ID: mdl-24955774

ABSTRACT

Chemotherapy-induced peripheral neuropathy (CIPN) characterized by loss of sensory sensitivity and pain in hands and feet is the major dose-limiting toxicity of many chemotherapeutics. At present, there are no FDA-approved treatments for CIPN. The anti-diabetic drug metformin is the most widely used prescription drug in the world and improves glycemic control in diabetes patients. There is some evidence that metformin enhances the efficacy of cancer treatment. The aim of this study was to test the hypothesis that metformin protects against chemotherapy-induced neuropathic pain and sensory deficits. Mice were treated with cisplatin together with metformin or saline. Cisplatin induced increased sensitivity to mechanical stimulation (mechanical allodynia) as measured using the von Frey test. Co-administration of metformin almost completely prevented the cisplatin-induced mechanical allodynia. Co-administration of metformin also prevented paclitaxel-induced mechanical allodynia. The capacity of the mice to detect an adhesive patch on their hind paw was used as a novel indicator of chemotherapy-induced sensory deficits. Co-administration of metformin prevented the cisplatin-induced increase in latency to detect the adhesive patch indicating that metformin prevents sensory deficits as well. Moreover, metformin prevented the reduction in density of intra-epidermal nerve fibers (IENFs) in the paw that develops as a result of cisplatin treatment. We conclude that metformin protects against pain and loss of tactile function in a mouse model of CIPN. The finding that metformin reduces loss of peripheral nerve endings indicates that mechanism underlying the beneficial effects of metformin includes a neuroprotective activity. Because metformin is widely used for treatment of type II diabetes, has a broad safety profile, and is currently being tested as an adjuvant drug in cancer treatment, clinical translation of these findings could be rapidly achieved.


Subject(s)
Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/drug therapy , Protective Agents/therapeutic use , Animals , Cisplatin/adverse effects , Disease Models, Animal , Hyperalgesia/chemically induced , Hyperalgesia/complications , Hyperalgesia/drug therapy , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Mice, Inbred C57BL , Nerve Fibers/drug effects , Nerve Fibers/pathology , Neuralgia/chemically induced , Neuralgia/complications , Neuralgia/drug therapy , Paclitaxel/adverse effects , Peripheral Nervous System Diseases/complications , Protective Agents/pharmacology
12.
J Neuroinflammation ; 9: 143, 2012 Jun 25.
Article in English | MEDLINE | ID: mdl-22731384

ABSTRACT

BACKGROUND: Chronic pain is often associated with microglia activation in the spinal cord. We recently showed that microglial levels of the kinase G protein-coupled receptor kinase (GRK)2 are reduced in models of chronic pain. We also found that mice with a cell-specific reduction of around 50% in GRK2 level in microglia/macrophages (LysM-GRK2+/- mice) develop prolonged inflammatory hyperalgesia concomitantly with ongoing spinal microglia/macrophage activation. The microRNA miR-124 is thought to keep microglia/macrophages in brain and spinal cord in a quiescent state. In the present study, we investigated the contribution of miR-124 to regulation of hyperalgesia and microglia/macrophage activation in GRK2-deficient mice. In addition, we investigated the effect of miR-124 on chronic inflammatory and neuropathic pain in wild-type (WT) mice. METHODS: Hyperalgesia was induced by intraplantar IL-1ß in WT and LysM-GRK2+/- mice. We determined spinal cord microglia/macrophage miR-124 expression and levels of pro-inflammatory M1 and anti-inflammatory M2 activation markers. The effect of intrathecal miR-124 treatment on IL-1ß-induced hyperalgesia and spinal M1/M2 phenotype, and on carrageenan-induced and spared nerve injury-induced chronic hyperalgesia in WT mice was analyzed. RESULTS: Transition from acute to persistent hyperalgesia in LysM-GRK2+/- mice is associated with reduced spinal cord microglia miR-124 levels. In our LysM-GRK2+/- mice, there was a switch towards a pro-inflammatory M1 phenotype together with increased pro-inflammatory cytokine production. Intrathecal administration of miR-124 completely prevented the transition to persistent pain in response to IL-1ß in LysM-GRK2+/- mice. The miR-124 treatment also normalized expression of spinal M1/M2 markers of LysM-GRK2+/- mice. Moreover, intrathecal miR-124 treatment reversed the persistent hyperalgesia induced by carrageenan in WT mice and prevented development of mechanical allodynia in the spared nerve injury model of chronic neuropathic pain in WT mice. CONCLUSIONS: We present the first evidence that intrathecal miR-124 treatment can be used to prevent and treat persistent inflammatory and neuropathic pain. In addition, we show for the first time that persistent hyperalgesia in GRK2-deficient mice is associated with an increased ratio of M1/M2 type markers in spinal cord microglia/macrophages, which is restored by miR-124 treatment. We propose that intrathecal miR-124 treatment might be a powerful novel treatment for pathological chronic pain with persistent microglia activation.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/deficiency , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , MicroRNAs/therapeutic use , Animals , Female , G-Protein-Coupled Receptor Kinase 2/genetics , Hyperalgesia/genetics , Injections, Spinal , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/administration & dosage , MicroRNAs/genetics , Microglia/drug effects , Microglia/metabolism , Treatment Outcome
13.
J Surg Oncol ; 106(1): 57-61, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22237993

ABSTRACT

BACKGROUND AND OBJECTIVE: High-mobility group box 1 (HMGB1) is a versatile protein with intranuclear and extracellular functions that is involved in numerous biological and pathological processes, such as transcription, DNA repair, and response to infection and inflammation. HMGB1 overexpression has been reported in a variety of human cancers. However, the clinical significance of HMGB1 expression in bladder cancer (BC) remains unclear. This study is aimed to investigate the correlations between HMGB1 expression and prognosis in patients with BC. METHODS: HMGB1 protein expression in 164 primary BC tissue specimens was analyzed by immunohistochemistry, and its association with clinicopathologic factors and prognosis was also analyzed. RESULTS: HMGB1 protein had high expression in 87 of 164 cases of BC (53%). HMGB1 overexpression was significantly associated with tumor grade (P < 0.001), and stage (P = 0.001). The Kaplan-Meier survival analysis demonstrated that HMGB1 expression was significantly associated with shorter disease-free survival and overall survival (both P < 0.001). Multivariate analysis further demonstrated that HMGB1 was an independent prognostic factor for patients with BC. CONCLUSIONS: HMGB1 might be a new molecular marker to predict the prognosis of patients with BC.


Subject(s)
Biomarkers, Tumor/analysis , Gene Expression Regulation, Neoplastic , HMGB1 Protein/analysis , Urinary Bladder Neoplasms/chemistry , Urinary Bladder Neoplasms/pathology , Adult , Aged , Cohort Studies , Disease-Free Survival , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Neoplasm Grading , Neoplasm Staging , Predictive Value of Tests , Prognosis , Proportional Hazards Models , Retrospective Studies , Up-Regulation , Urinary Bladder Neoplasms/mortality
SELECTION OF CITATIONS
SEARCH DETAIL
...