Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cancer Med ; 13(15): e70077, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39101490

RESUMEN

AIMS: Bone cancer produces severe pain that is treated with opioids, but serious side effects limit opioid utilization. There is therefore a need to develop effective and safe non-opioid alternatives. The lipid mediator, Resolvin D1 (RvD1), could be a prospective candidate for cancer pain treatment. To assess RvD1 and other potential candidates, appropriate animal models that recapitulate clinical features must be used. Although several preclinical models of cancer pain have been developed, the influence of sex on the development of cancer pain and the effectiveness of RvD1 have not been studied. RESULTS: Using a mouse model of fibrosarcoma growth in and around the calcaneus bone, we demonstrated that the mechanical hyperalgesia in the tumor-bearing hind paw develops independently of sex, except that it developed a little sooner in female mice. A single intravenous injection of RvD1 (0.001-10 µg/kg) decreased hyperalgesia in both sexes with similar potency (ED50 = 0.0015 µg/kg) and efficacy. Repeated daily administration of 10 µg/kg RvD1 prolonged the analgesic effect and completely abolished hyperalgesia. This was also independent of sex. CONCLUSION: In this preclinical mouse model of bone cancer pain, the development of pain and the analgesic effectiveness of RvD1 are not influenced by sex.


Asunto(s)
Neoplasias Óseas , Dolor en Cáncer , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos , Hiperalgesia , Animales , Femenino , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/complicaciones , Neoplasias Óseas/secundario , Masculino , Dolor en Cáncer/tratamiento farmacológico , Dolor en Cáncer/etiología , Ácidos Docosahexaenoicos/administración & dosificación , Ácidos Docosahexaenoicos/farmacología , Ratones , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/etiología , Analgésicos/farmacología , Analgésicos/administración & dosificación , Fibrosarcoma/tratamiento farmacológico , Fibrosarcoma/patología , Fibrosarcoma/complicaciones , Factores Sexuales , Dimensión del Dolor
2.
Mol Pain ; 20: 17448069241259535, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38773702

RESUMEN

Methylene blue (MB) has been shown to reduce mortality and morbidity in vasoplegic patients after cardiac surgery. Though MB is considered to be safe, extravasation of MB leading to cutaneous toxicity has been reported. In this study, we sought to characterize MB-induced cutaneous toxicity and investigate the underlying mechanisms. To induce MB-induced cutaneous toxicity, we injected 64 adult male Sprague-Dawley rates with 200 µL saline (vehicle) or 1%, 0.1%, or 0.01% MB in the plantar hind paws. Paw swelling, skin histologic changes, and heat and mechanical hyperalgesia were measured. Injection of 1%, but not 0.1% or 0.01% MB, produced significant paw swelling compared to saline. Injection of 1% MB produced heat hyperalgesia but not mechanical hyperalgesia. Pain behaviors were unchanged following injections of 0.1% or 0.01% MB. Global transcriptomic analysis by RNAseq identified 117 differentially expressed genes (111 upregulated, 6 downregulated). Ingenuity Pathway Analysis showed an increased quantity of leukocytes, increased lipids, and decreased apoptosis of myeloid cells and phagocytes with activation of IL-1ß and Fos as the two major regulatory hubs. qPCR showed a 16-fold increase in IL-6 mRNA. Thus, using a novel rat model of MB-induced cutaneous toxicity, we show that infiltration of 1% MB into cutaneous tissue causes a dose-dependent pro-inflammatory response, highlighting potential roles of IL-6, IL-1ß, and Fos. Thus, anesthesiologists should administer dilute MB intravenously through peripheral venous catheters. Higher concentrations of MB (1%) should be administered through a central venous catheter to minimize the risk of cutaneous toxicity.


Asunto(s)
Modelos Animales de Enfermedad , Hiperalgesia , Inflamación , Azul de Metileno , Ratas Sprague-Dawley , Piel , Animales , Masculino , Azul de Metileno/farmacología , Azul de Metileno/administración & dosificación , Hiperalgesia/patología , Hiperalgesia/inducido químicamente , Inflamación/patología , Inflamación/inducido químicamente , Piel/efectos de los fármacos , Piel/patología , Relación Dosis-Respuesta a Droga , Calor , Ratas , Interleucina-1beta/metabolismo , Interleucina-1beta/genética
3.
Blood ; 142(22): 1918-1927, 2023 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-37774369

RESUMEN

Vaso-occlusive pain episodes (VOE) cause severe pain in patients with sickle cell disease (SCD). Vaso-occlusive events promote ischemia/reperfusion pathobiology that activates complement. We hypothesized that complement activation is linked to VOE. We used cold to induce VOE in the Townes sickle homozygous for hemoglobin S (HbSS) mouse model and complement inhibitors to determine whether anaphylatoxin C5a mediates VOE. We used a dorsal skinfold chamber to measure microvascular stasis (vaso-occlusion) and von Frey filaments applied to the plantar surface of the hind paw to assess mechanical hyperalgesia in HbSS and control Townes mice homozygous for hemoglobin A (HbAA) mice after cold exposure at 10°C/50°F for 1 hour. Cold exposure induced more vaso-occlusion in nonhyperalgesic HbSS mice (33%) than in HbAA mice (11%) or HbSS mice left at room temperature (1%). Cold exposure also produced mechanical hyperalgesia as measured by paw withdrawal threshold in HbSS mice compared with that in HbAA mice or HbSS mice left at room temperature. Vaso-occlusion and hyperalgesia were associated with an increase in complement activation fragments Bb and C5a in plasma of HbSS mice after cold exposure. This was accompanied by an increase in proinflammatory NF-κB activation and VCAM-1 and ICAM-1 expression in the liver. Pretreatment of nonhyperalgesic HbSS mice before cold exposure with anti-C5 or anti-C5aR monoclonal antibodies (mAbs) decreased vaso-occlusion, mechanical hyperalgesia, complement activation, and liver inflammatory markers compared with pretreatment with control mAb. Anti-C5 or -C5aR mAb infusion also abrogated mechanical hyperalgesia in HbSS mice with ongoing hyperalgesia at baseline. These findings suggest that C5a promotes vaso-occlusion, pain, and inflammation during VOE and may play a role in chronic pain.


Asunto(s)
Anemia de Células Falciformes , Rasgo Drepanocítico , Ratones , Humanos , Animales , Hiperalgesia/etiología , Hiperalgesia/metabolismo , Ratones Transgénicos , Dolor , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/metabolismo , Rasgo Drepanocítico/complicaciones , Activación de Complemento
4.
Pain Rep ; 8(5): e1097, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37711430

RESUMEN

Background: This study investigated if a localized increase in skin temperature in rat models of incisional and inflammatory pain correlates with the intensity of spontaneous and evoked pain behaviors. Methods: Anesthetized rats received either a 20-mm longitudinal incision made through the skin, fascia, and muscle of the plantar hind paw or an injection of complete Freund adjuvant into the plantar hind paw of anesthetized rats to induce local inflammation. Spontaneous and evoked pain behaviors were assessed, and changes in skin temperature were measured using a noncontact infrared thermometer. Results: There were no differences in skin temperature between the ipsilateral and contralateral hind paw before the incision or inflammation. Skin temperature increased at 2 hours after hind paw plantar incision or 1 day after inflammation of the affected paw, which gradually returned to baseline by the first day and fourth days after treatment, respectively. The increase in skin temperature correlated with the intensity of spontaneous pain behaviors and heat but not with mechanical allodynia. Conclusions: Our results suggest that a simple measurement of localized skin temperature using a noncontact infrared thermometer could measure the extent of spontaneous pain behaviors and heat hyperalgesia following plantar incision or inflammation in animals. In the absence of a reliable objective marker of pain, these results are encouraging. However, studies are warranted to validate our results using analgesics and pain-relieving interventions, such as nerve block on skin temperature changes.

5.
Pain ; 164(12): 2684-2695, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37278638

RESUMEN

ABSTRACT: Pain associated with bone cancer remains poorly managed, and chemotherapeutic drugs used to treat cancer usually increase pain. The discovery of dual-acting drugs that reduce cancer and produce analgesia is an optimal approach. The mechanisms underlying bone cancer pain involve interactions between cancer cells and nociceptive neurons. We demonstrated that fibrosarcoma cells express high levels of autotaxin (ATX), the enzyme synthetizing lysophosphatidic acid (LPA). Lysophosphatidic acid increased proliferation of fibrosarcoma cells in vitro. Lysophosphatidic acid is also a pain-signaling molecule, which activates LPA receptors (LPARs) located on nociceptive neurons and satellite cells in dorsal root ganglia. We therefore investigated the contribution of the ATX-LPA-LPAR signaling to pain in a mouse model of bone cancer pain in which fibrosarcoma cells are implanted into and around the calcaneus bone, resulting in tumor growth and hypersensitivity. LPA was elevated in serum of tumor-bearing mice, and blockade of ATX or LPAR reduced tumor-evoked hypersensitivity. Because cancer cell-secreted exosomes contribute to hypersensitivity and ATX is bound to exosomes, we determined the role of exosome-associated ATX-LPA-LPAR signaling in hypersensitivity produced by cancer exosomes. Intraplantar injection of cancer exosomes into naive mice produced hypersensitivity by sensitizing C-fiber nociceptors. Inhibition of ATX or blockade of LPAR attenuated cancer exosome-evoked hypersensitivity in an ATX-LPA-LPAR-dependent manner. Parallel in vitro studies revealed the involvement of ATX-LPA-LPAR signaling in direct sensitization of dorsal root ganglion neurons by cancer exosomes. Thus, our study identified a cancer exosome-mediated pathway, which may represent a therapeutic target for treating tumor growth and pain in patients with bone cancer.


Asunto(s)
Neoplasias Óseas , Dolor en Cáncer , Exosomas , Fibrosarcoma , Humanos , Animales , Ratones , Dolor en Cáncer/etiología , Lisofosfolípidos/metabolismo , Neoplasias Óseas/complicaciones , Dolor/tratamiento farmacológico , Dolor/etiología
6.
Neuroscience ; 526: 1-12, 2023 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-37330194

RESUMEN

Sickle cell disease (SCD) is an inherited blood disorder that is associated with acute episodic and chronic pain. Mice with SCD have robust hyperalgesia mediated, in part, by sensitization of spinal dorsal horn neurons. However, underlying mechanisms are not fully understood. Since the rostral ventromedial medulla (RVM) is a major component of descending circuitry that modulates nociceptive transmission in the spinal cord, we examined if the RVM contributes to hyperalgesia in mice with SCD. Injection of lidocaine, but not vehicle, into the RVM eliminated mechanical and heat hyperalgesia in sickle (HbSS-BERK) mice without altering mechanical and heat sensitivity in naïve C57B mice. These data indicate that the RVM contributes to the maintenance of hyperalgesia in mice with SCD. In electrophysiological studies, we determined the changes in response properties of RVM neurons that might contribute to hyperalgesia in sickle mice. Recordings were made from single ON, OFF, and Neutral cells in the RVM of sickle and control (HbAA-BERK) mice. Spontaneous activity and responses of ON, OFF and Neutral cells evoked by heat (50 °C) and mechanical (26 g) stimuli applied to the hind paw were compared between sickle and control mice. Although there were no differences in the proportions of functionally-identified neurons or spontaneous activity between sickle and control mice, evoked responses of ON cells to heat and mechanical stimuli were increased approximately 3-fold in sickle mice as compared to control mice. Thus, the RVM contributes to hyperalgesia in sickle mice via a specific ON cell-dependent descending facilitation of nociceptive transmission.


Asunto(s)
Anemia de Células Falciformes , Hiperalgesia , Ratas , Ratones , Animales , Nocicepción/fisiología , Ratas Sprague-Dawley , Neuronas/fisiología , Anemia de Células Falciformes/complicaciones , Bulbo Raquídeo
7.
Neuroscience ; 516: 54-61, 2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-36805004

RESUMEN

MMG22 is a bivalent ligand containing MOR agonist and mGluR5 antagonist pharmacophores connected by a 22-atom linker. Intrathecal (i.t.) administration of MMG22 to inflamed mice has been reported to produce fmol-range antinociception in the reversal of LPS-induced hyperalgesia. MMG22 reduced hyperalgesia in the spared nerve injury (SNI) model of neuropathic pain at 10 days after injury but not at 30 days after injury, perhaps related to the inflammation that occurs early after injury but subsequently subsides. The present study determined the efficacy of MMG22 in cisplatin-treated male mice in order to provide data relating to the efficacy of MMG22 in the treatment of neuropathic pain that is associated with inflammation. Groups of eight mice each received daily intraperitoneal (i.p.) injections of cisplatin for seven days to produce robust mechanical allodynia defined by the decrease in withdrawal threshold using an electronic von Frey applied to the plantar surface of the hind paw. Intrathecal administration of MMG22 potently reduced mechanical hyperalgesia (ED50 0.04 fmol/mouse) without tolerance, whereas MMG10 was essentially inactive. Morphine was less potent than MMG22 by >5-orders of magnitude and displayed tolerance. Subcutaneous MMG22 was effective (ED50 = 2.41 mg/kg) and devoid of chronic tolerance. We propose that MMG22 induces the formation of a MOR-mGluR5 heteromer through selective interaction with the upregulated NR2B subunit of activated NMDAR, in view of the 4600-fold reduction of i.t. MMG22 antinociception by the selective NR2B antagonist, Ro25-6981. A possible explanation for the substantially reduced potency for MMG22 in the SNI model is discussed.


Asunto(s)
Hiperalgesia , Neuralgia , Ratones , Masculino , Animales , Hiperalgesia/tratamiento farmacológico , Cisplatino , Morfina/farmacología , Neuralgia/inducido químicamente , Neuralgia/tratamiento farmacológico , Inflamación , Modelos Animales de Enfermedad
8.
Haematologica ; 108(3): 859-869, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35615929

RESUMEN

Sickle cell disease (SCD) is the most common inherited disease. Pain is a key morbidity of SCD and opioids are the main treatment but their side effects emphasize the need for new analgesic approaches. Humanized transgenic mouse models have been instructive in understanding the pathobiology of SCD and mechanisms of pain. Homozygous (HbSS) Berkley mice express >99% human sickle hemoglobin and several features of clinical SCD including hyperalgesia. Previously, we reported that the endocannabinoid 2-arachidonoylglycerol (2-AG) is a precursor of the pro-nociceptive mediator prostaglandin E2-glyceryl ester (PGE2-G) which contributes to hyperalgesia in SCD. We now demonstrate the causal role of 2-AG in hyperalgesia in sickle mice. Hyperalgesia in HbSS mice correlated with elevated levels of 2-AG in plasma, its synthesizing enzyme diacylglycerol lipase ß (DAGLß) in blood cells, and with elevated levels of PGE2 and PGE2-G, pronociceptive derivatives of 2-AG. A single intravenous injection of 2-AG produced hyperalgesia in non-hyperalgesic HbSS mice, but not in control (HbAA) mice expressing normal human HbA. JZL184, an inhibitor of 2-AG hydrolysis, also produced hyperalgesia in non-hyperalgesic HbSS or hemizygous (HbAS) mice, but did not influence hyperalgesia in hyperalgesic HbSS mice. Systemic and intraplantar administration of KT109, an inhibitor of DAGLß, decreased mechanical and heat hyperalgesia in HbSS mice. The decrease in hyperalgesia was accompanied by reductions in 2-AG, PGE2 and PGE2-G in the blood. These results indicate that maintaining the physiological level of 2-AG in the blood by targeting DAGLß may be a novel and effective approach to treat pain in SCD.


Asunto(s)
Anemia de Células Falciformes , Hiperalgesia , Ratones , Humanos , Animales , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/etiología , Dinoprostona , Dolor/tratamiento farmacológico , Dolor/etiología , Ratones Transgénicos , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/tratamiento farmacológico , Hemoglobina Falciforme
9.
Mol Pain ; 18: 17448069221142523, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-36408567

RESUMEN

Methylene blue (MB) is an effective treatment for methemoglobinemia, ifosfamide-induced encephalopathy, cyanide poisoning, and refractory vasoplegia. However, clinical case reports and preclinical studies indicate potentially neurotoxic activity of MB at certain concentrations. The exact mechanisms of MB neurotoxicity are not known, and while the effects of MB on neuronal tissue from different brain regions and myenteric ganglia have been examined, its effects on primary afferent neurons from dorsal root ganglia (DRG) have not been studied. Mouse DRG were exposed to MB (0.3-10 µM) in vitro to assess neurite outgrowth. Increasing concentrations of MB (0.3-10 µM) were associated with neurotoxicity as shown by a substantial loss of cells with neurite formation, particularly at 10 µM. In parallel experiments, cultured rat DRG neurons were treated with MB (100 µM) to examine how MB affects electrical membrane properties of small-diameter sensory neurons. MB decreased peak inward and outward current densities, decreased action potential amplitude, overshoot, afterhyperpolarization, increased action potential rise time, and decreased action potential firing in response to current stimulation. MB induced dose-dependent toxicity in peripheral neurons, in vitro. These findings are consistent with studies in brain and myenteric ganglion neurons showing increased neuronal loss and altered membrane electrical properties after MB application. Further research is needed to parse out the toxicity profile for MB to minimize damage to neuronal structures and reduce side effects in clinical settings.


Asunto(s)
Ganglios Espinales , Azul de Metileno , Ratas , Ratones , Animales , Azul de Metileno/farmacología , Azul de Metileno/metabolismo , Ganglios Espinales/metabolismo , Células Receptoras Sensoriales/metabolismo , Electrofisiología , Técnicas de Cultivo de Célula , Células Cultivadas
11.
Neurosci Lett ; 753: 135845, 2021 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-33774149

RESUMEN

Although millions of people are diagnosed with cancer each year, survival has never been greater thanks to early diagnosis and treatments. Powerful chemotherapeutic agents are highly toxic to cancer cells, but because they typically do not target cancer cells selectively, they are often toxic to other cells and produce a variety of side effects. In particular, many common chemotherapies damage the peripheral nervous system and produce neuropathy that includes a progressive degeneration of peripheral nerve fibers. Chemotherapy-induced peripheral neuropathy (CIPN) can affect all nerve fibers, but sensory neuropathies are the most common, initially affecting the distal extremities. Symptoms include impaired tactile sensitivity, tingling, numbness, paraesthesia, dysesthesia, and pain. Since neuropathic pain is difficult to manage, and because degenerated nerve fibers may not grow back and regain normal function, considerable research has focused on understanding how chemotherapy causes painful CIPN so it can be prevented. Due to the fact that both therapeutic and side effects of chemotherapy are primarily associated with the accumulation of reactive oxygen species (ROS) and oxidative stress, this review focuses on the activation of endogenous antioxidant pathways, especially PPARγ, in order to prevent the development of CIPN and associated pain. The use of synthetic and natural PPARγ agonists to prevent CIPN is discussed.


Asunto(s)
Antineoplásicos/efectos adversos , Neoplasias/tratamiento farmacológico , Neuralgia/prevención & control , PPAR gamma/agonistas , Animales , Modelos Animales de Enfermedad , Humanos , Neuralgia/inducido químicamente , Estrés Oxidativo/efectos de los fármacos , PPAR gamma/metabolismo , Especies Reactivas de Oxígeno/metabolismo
12.
Neuroscience ; 457: 74-87, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33422618

RESUMEN

Pain is a major health problem, affecting over fifty million adults in the US alone, with significant economic cost in medical care and lost productivity. Despite evidence implicating nicotinic acetylcholine receptors (nAChRs) in pathological pain, their specific contribution to pain processing in the spinal cord remains unclear given their presence in both neuronal and non-neuronal cell types. Here we investigated if loss of neuronal-specific TMEM35a (NACHO), a novel chaperone for functional expression of the homomeric α7 and assembly of the heteromeric α3, α4, and α6-containing nAChRs, modulates pain in mice. Mice with tmem35a deletion exhibited thermal hyperalgesia and mechanical allodynia. Intrathecal administration of nicotine and the α7-specific agonist, PHA543613, produced analgesic responses to noxious heat and mechanical stimuli in tmem35a KO mice, respectively, suggesting residual expression of these receptors or off-target effects. Since NACHO is expressed only in neurons, these findings indicate that neuronal α7 nAChR in the spinal cord contributes to heat nociception. To further determine the molecular basis underlying the pain phenotype, we analyzed the spinal cord transcriptome. Compared to WT control, the spinal cord of tmem35a KO mice exhibited 72 differentially-expressed genes (DEGs). These DEGs were mapped onto functional gene networks using the knowledge-based database, Ingenuity Pathway Analysis, and suggests increased neuroinflammation as a potential contributing factor for the hyperalgesia in tmem35a KO mice. Collectively, these findings implicate a heightened inflammatory response in the absence of neuronal NACHO activity. Additional studies are needed to determine the precise mechanism by which NACHO in the spinal cord modulates pain.


Asunto(s)
Hiperalgesia , Receptores Nicotínicos , Animales , Canales Iónicos , Ratones , Chaperonas Moleculares/metabolismo , Neuronas/metabolismo , Nicotina , Receptores Nicotínicos/genética
13.
Mol Pain ; 16: 1744806920956480, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32909881

RESUMEN

To develop non-opioid therapies for postoperative incisional pain, we must understand its underlying molecular mechanisms. In this study, we assessed global gene expression changes in dorsal root ganglia neurons in a model of incisional pain to identify pertinent molecular pathways. Male, Sprague-Dawley rats underwent infiltration of 1% capsaicin or vehicle into the plantar hind paw (n = 6-9/group) 30 min before plantar incision. Twenty-four hours after incision or sham (control) surgery, lumbar L4-L6 dorsal root ganglias were collected from rats pretreated with vehicle or capsaicin. RNA was isolated and sequenced by next generation sequencing. The genes were then annotated to functional networks using a knowledge-based database, Ingenuity Pathway Analysis. In rats pretreated with vehicle, plantar incision caused robust hyperalgesia, up-regulated 36 genes and downregulated 90 genes in dorsal root ganglias one day after plantar incision. Capsaicin pretreatment attenuated pain behaviors, caused localized denervation of the dermis and epidermis, and prevented the incision-induced changes in 99 of 126 genes. The pathway analyses showed altered gene networks related to increased pro-inflammatory and decreased anti-inflammatory responses in dorsal root ganglias. Insulin-like growth factor signaling was identified as one of the major gene networks involved in the development of incisional pain. Expression of insulin-like growth factor -2 and IGFBP6 in dorsal root ganglia were independently validated with quantitative real-time polymerase chain reaction. We discovered a distinct subset of dorsal root ganglia genes and three key signaling pathways that are altered 24 h after plantar incision but are unchanged when incision was made after capsaicin infiltration in the skin. Further exploration of molecular mechanisms of incisional pain may yield novel therapeutic targets.


Asunto(s)
Capsaicina/farmacología , Ganglios Espinales/metabolismo , Dolor Postoperatorio/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Somatomedinas/metabolismo , Transcriptoma/genética , Animales , Escala de Evaluación de la Conducta , Capsaicina/uso terapéutico , Biología Computacional , Regulación hacia Abajo , Ganglios Espinales/lesiones , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/metabolismo , Masculino , RNA-Seq , Ratas , Ratas Sprague-Dawley , Transducción de Señal/genética , Somatomedinas/genética , Herida Quirúrgica/complicaciones , Regulación hacia Arriba
14.
Neuroscience ; 446: 102-112, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32858141

RESUMEN

It was recently shown that local injection, systemic administration or topical application of the peripherally-restricted mu-opioid receptor (MOR) agonist loperamide (Lo) and the delta-opioid receptor (DOR) agonist oxymorphindole (OMI) synergized to produce highly potent anti-hyperalgesia that was dependent on both MOR and DOR located in the periphery. We assessed peripheral mechanisms by which this Lo/OMI combination produces analgesia in mice expressing the light-sensitive protein channelrhodopsin2 (ChR2) in neurons that express NaV1.8 voltage-gated sodium channels. These mice (NaV1.8-ChR2+) enabled us to selectively target and record electrophysiological activity from these neurons (the majority of which are nociceptive) using blue light stimulation of the hind paw. We assessed the effect of Lo/OMI on nociceptor activity in both naïve mice and mice treated with complete Freund's adjuvant (CFA) to induce chronic inflammation of the hind paw. Teased fiber recording of tibial nerve fibers innervating the plantar hind paw revealed that the Lo/OMI combination reduced responses to light stimulation in naïve mice and attenuated spontaneous activity (SA) as well as responses to light and mechanical stimuli in CFA-treated mice. These results show that Lo/OMI reduces activity of C-fiber nociceptors that express NaV1.8 and corroborate recent behavioral studies demonstrating the potent analgesic effects of this drug combination. Because of its peripheral site of action, Lo/OMI might produce effective analgesia without the side effects associated with activation of opioid receptors in the central nervous system.


Asunto(s)
Loperamida , Nociceptores , Animales , Hiperalgesia/tratamiento farmacológico , Inflamación , Loperamida/farmacología , Ratones , Morfolinas , Fibras Nerviosas Amielínicas , Receptores Opioides delta , Receptores Opioides mu
15.
Prostaglandins Other Lipid Mediat ; 151: 106479, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32745525

RESUMEN

Pain produced by bone cancer is often severe and difficult to treat. Here we examined effects of Resolvin D1 (RvD1) or E1 (RvE1), antinociceptive products of ω-3 polyunsaturated fatty acids, on cancer-induced mechanical allodynia and heat hyperalgesia. Experiments were performed using a mouse model of bone cancer produced by implantation of osteolytic ficrosarcoma into and around the calcaneus bone. Mechanical allodynia and heat hyperalgesia in the tumor-bearing paw were assessed by measuring withdrawal responses to a von Frey monofilament and to radiant heat applied on the plantar hind paw. RvD1, RvE1, and cannabinoid receptor antagonists were injected intrathecally. Spinal content of endocannabinoids was evaluated using UPLC-MS/MS analysis. RvD1 and RvE1 had similar antinociceptive potencies. ED50s for RvD1 and RvE1 in reducing mechanical allodynia were 0.2 pg (0.53 fmol) and 0.6 pg (1.71 fmol), respectively, and were 0.3 pg (0.8 fmol) and 0.2 pg (0.57 fmol) for reducing heat hyperalgesia. Comparisons of dose-response relationships showed equal efficacy for reducing mechanical allodynia, however, efficacy for reducing heat hyperalgesia was greater for of RvD1. Using UPLC-MS/MS we determined that RvD1, but not RvE1, increased levels of the endocannabinoids Anandamide and 2-Arachidonoylglycerol in the spinal cord. Importantly, Resolvins did not alter acute nociception or motor function in naïve mice. Our data indicate, that RvD1 and RvE1 produce potent antiallodynia and antihyperalgesia in a model of bone cancer pain. RvD1 also triggers spinal upregulation of endocannabinoids that produce additional antinociception predominantly through CB2 receptors.


Asunto(s)
Neoplasias Óseas/complicaciones , Dolor en Cáncer/tratamiento farmacológico , Ácidos Docosahexaenoicos/farmacología , Ácido Eicosapentaenoico/análogos & derivados , Endocannabinoides/metabolismo , Hiperalgesia/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Animales , Dolor en Cáncer/patología , Ácidos Docosahexaenoicos/uso terapéutico , Ácido Eicosapentaenoico/farmacología , Ácido Eicosapentaenoico/uso terapéutico , Hiperalgesia/patología , Masculino , Ratones
16.
Pain ; 161(9): 2041-2057, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32345918

RESUMEN

ABSTRACT: Functional interactions between the mu opioid receptor (MOR) and the metabotropic glutamate receptor 5 (mGluR5) in pain and analgesia have been well established. MMG22 is a bivalent ligand containing MOR agonist (oxymorphamine) and mGluR5 antagonist (MPEP) pharmacophores tethered by a 22-atom linker. MMG22 has been shown to produce potent analgesia in several models of chronic inflammatory and neuropathic pain (NP). This study assessed the efficacy of systemic administration of MMG22 at reducing pain behavior in the spared nerve injury (SNI) model of NP in mice, as well as its side-effect profile and abuse potential. MMG22 reduced mechanical hyperalgesia and spontaneous ongoing pain after SNI, with greater potency early (10 days) as compared to late (30 days) after injury. Systemic administration of MMG22 did not induce place preference in naive animals, suggesting absence of abuse liability when compared to traditional opioids. MMG22 also lacked the central locomotor, respiratory, and anxiolytic side effects of its monomeric pharmacophores. Evaluation of mRNA expression showed the transcripts for both receptors were colocalized in cells in the dorsal horn of the lumbar spinal cord and dorsal root ganglia. Thus, MMG22 reduces hyperalgesia after injury in the SNI model of NP without the typical centrally mediated side effects associated with traditional opioids.


Asunto(s)
Analgésicos Opioides , Neuralgia , Analgésicos Opioides/uso terapéutico , Animales , Hiperalgesia/tratamiento farmacológico , Ligandos , Ratones , Neuralgia/tratamiento farmacológico , Receptor del Glutamato Metabotropico 5 , Receptores Opioides mu/genética
17.
J Neurophysiol ; 123(5): 1944-1954, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32292106

RESUMEN

Understanding of cortical encoding of itch is limited. Injection of pruritogens and algogens into the skin of the cheek produces distinct behaviors, making the rodent cheek a useful model for understanding mechanisms of itch and pain. We examined responses of neurons in the primary somatosensory cortex by application of mechanical stimuli (brush, pressure, and pinch) and stimulations with intradermal injections of pruritic and algesic chemical of receptive fields located on the skin of the cheek in urethane-anesthetized rats. Stimuli included chloroquine, serotonin, ß-alanine, histamine, capsaicin, and mustard oil. All 33 neurons studied were excited by noxious mechanical stimuli applied to the cheek. Based on mechanical stimulation most neurons were functionally classified as high threshold. Of 31 neurons tested for response to chemical stimuli, 84% were activated by one or more pruritogens/partial pruritogens. No cells were activated by all five substances. Histamine activated the greatest percentage of neurons and evoked the greatest mean discharge. Importantly, no cells were excited exclusively by pruritogens or partial pruritogens. The recording sites of all neurons that responded to chemical stimuli applied to the cheek were located in the dysgranular zone (DZ) and in deep laminae of the medial border of the vibrissal barrel fields (VBF). Therefore, neurons in the DZ/VBF of rats encode mechanical and chemical pruritogens and algogens. This cortical region appears to contain primarily nociceptive neurons as defined by responses to noxious pinching of the skin. Its role in encoding itch and pain from the cheek of the face needs further study.NEW & NOTEWORTHY Processing of information related to itch sensation at the level of cerebral cortex is not well understood. In this first single-unit electrophysiological study of pruriceptive cortical neurons, we show that neurons responsive to noxious and pruritic stimulation of the cheek of the face are concentrated in a small area of the dysgranular cortex, indicating that these neurons encode information related to itch and pain.


Asunto(s)
Fenómenos Electrofisiológicos/fisiología , Neuronas/fisiología , Nocicepción/fisiología , Prurito/fisiopatología , Corteza Somatosensorial/fisiopatología , Animales , Modelos Animales de Enfermedad , Inyecciones Intradérmicas , Masculino , Estimulación Física , Prurito/inducido químicamente , Prurito/etiología , Ratas , Ratas Sprague-Dawley
18.
Eur J Pain ; 24(6): 1197-1208, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32219920

RESUMEN

BACKGROUND: Spontaneous pain after surgical incision is a significant problem for most post-operative patients. Pain management that relies on opioids is hindered by numerous side effects, fuelling interest in non-opioid alternatives and multimodal approaches. Subcutaneous capsaicin infiltration has shown potential for reducing post-operative pain, but there are unanswered questions about safety and possible side effects. In adult rats, we characterized the analgesic effects of pre-operative capsaicin infiltration into the skin prior to plantar incision and assessed wound healing and epidermal innervation. METHODS: The surgical site on the plantar surface of the rat hind paw was infiltrated with 1% capsaicin or vehicle 30 min or 1 week prior to surgical incision. Spontaneous and evoked pain behaviours were assessed. Digital images of incised hind paws were used to quantify the surface area of the wound after suture removal. Epidermal nerve fibre quantification was performed on peri-incisional tissue biopsies. RESULTS: Intraplantar administration of capsaicin 30 min before surgical incision attenuated spontaneous pain behaviours, heat hyperalgesia, epidermal innervation, but it did not alter the rate of wound healing. Incisional pain hypersensitivity returned to baseline 2 weeks post-incision, at a time when no recovery of epidermal innervation is observed. CONCLUSIONS: Subcutaneous infiltration of capsaicin prior to surgical incision attenuated incision-induced pain behaviours and reduced epidermal innervation around the incision site. The long-lasting epidermal denervation by capsaicin had no impact in the rate of wound healing and recovery from pain behaviours. SIGNIFICANCE: Pre-operative capsaicin infiltration attenuated spontaneous pain-like behaviour and prevented the development of heat hyperalgesia following plantar skin incision. While capsaicin caused long-lasting and widespread loss of epidermal and dermal nerve fibres, there was no measurable impact on the rate of wound healing. Pre- or intra-operative infiltration of capsaicin into surgical sites could act as a safe prophylactic for post-operative pain and reduce the need for opioids during recovery.


Asunto(s)
Capsaicina , Hiperalgesia , Animales , Capsaicina/farmacología , Desnervación , Humanos , Hiperalgesia/tratamiento farmacológico , Microscopía Confocal , Dolor Postoperatorio/tratamiento farmacológico , Ratas
19.
Pain Med ; 21(1): 109-117, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31268147

RESUMEN

OBJECTIVES: Intrathecal baclofen (ITB) pumps used to manage spasticity in children with cerebral palsy (CP) also improve pain outcomes for some but not all patients. The purpose of this clinical feasibility study was to explore whether a quantitative sensory testing approach could a) be modified and used to subgroup individuals into sensory profiles and b) test whether the profiles were related to postimplant pain outcomes (i.e., pain responsive or pain persistent). SUBJECTS: A purposeful clinical sample of nine children with CP (mean age = 12.5 years, male = 56%) and complex communication needs participated. METHODS: A prospective within-subject design was used to measure proxy-reported pain before and after ITB implant. Pain response status was determined by proxy-reported pain intensity change (>50% change in maximum rated intensity). A modified quantitative sensory testing (mQST) procedure was used to assess behavioral responsivity to an array of calibrated sensory (tactile/acute nociceptive) stimuli before surgery. RESULTS: Seven individuals with presurgical pain had mQST differentiated sensory profiles in relation to ITB pain outcomes and relative to the two individuals with no pain. Presurgically, the ITB pain responsive subgroup (N = 3, maximum rated pain intensity decreased >50% after ITB implant) showed increased behavioral reactivity to an acute nociceptive stimulus and cold stimulus, whereas the ITB pain persistent subgroup (N = 4) showed reduced behavioral reactivity to cold and repeated von Frey stimulation relative to the no pain individuals. CONCLUSION: Implications for patient selection criteria and stratification to presurgically identify individuals with CP "at risk" for persistent postprocedure pain are discussed.


Asunto(s)
Baclofeno/administración & dosificación , Parálisis Cerebral/tratamiento farmacológico , Relajantes Musculares Centrales/administración & dosificación , Dolor/diagnóstico , Estimulación Física , Adolescente , Adulto , Parálisis Cerebral/complicaciones , Niño , Estudios de Factibilidad , Femenino , Humanos , Bombas de Infusión Implantables , Inyecciones Espinales , Masculino , Espasticidad Muscular/tratamiento farmacológico , Espasticidad Muscular/etiología , Dolor/etiología , Estimulación Física/instrumentación , Estimulación Física/métodos , Sensación/efectos de los fármacos , Adulto Joven
20.
Neuropharmacology ; 160: 107690, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31271770

RESUMEN

Pain is among the most common symptoms in cancer and approximately 90% of patients experience end-stage cancer pain. The management of cancer pain is challenging due to the significant side effects associated with opioids, and novel therapeutic approaches are needed. MMG22 is a bivalent ligand containing MOR agonist and mGluR5 antagonist pharmacophores joined by a 22-atom spacer. MMG22 exhibited extraordinary analgesia following intrathecal administration in a mouse model of bone cancer pain. Here, we assessed the effectiveness of systemic administration of MMG22 in reducing cancer pain and evaluated whether MMG22 displays side effects associated with opioids. Fibrosarcoma cells were injected into and around the calcaneus bone in C3H mice. Mechanical hyperalgesia was defined as an increase in the paw withdrawal frequencies (PWFs) evoked by application of a von Frey monofilament (3.9 mN bending force) applied to the plantar surface of the hind paw Subcutaneous (s.c.), intramuscular (i.m.), and oral (p.o.) administration of MMG22 produced robust dose-dependent antihyperalgesia, whose ED50 was orders of magnitude lower than morphine. Moreover, the ED50 for MMG22 decreased with disease progression. Importantly, s.c. administration of MMG22 did not produce acute (24 h) or long-term (9 days) tolerance, was not rewarding (conditioned place preference test), and did not produce naloxone-induced precipitated withdrawal or alter motor function. A possible mechanism of action of MMG22 is discussed in terms of inhibition of spinal NMDAR via antagonism of its co-receptor, mGluR5, and concomitant activation of neuronal MOR. We suggest that MMG22 may be a powerful alternative to traditional opioids for managing cancer pain. This article is part of the Special Issue entitled 'New Vistas in Opioid Pharmacology'.


Asunto(s)
Dolor en Cáncer/tratamiento farmacológico , Dolor en Cáncer/metabolismo , Receptores de Ácido Kaínico/antagonistas & inhibidores , Receptores Opioides mu/agonistas , Animales , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/metabolismo , Modelos Animales de Enfermedad , Vías de Administración de Medicamentos , Fibrosarcoma/tratamiento farmacológico , Fibrosarcoma/metabolismo , Hiperalgesia/tratamiento farmacológico , Ligandos , Masculino , Ratones , Ratones Endogámicos C3H , Morfina/uso terapéutico , Receptores de Ácido Kaínico/administración & dosificación , Receptores Opioides mu/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...