Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Front Integr Neurosci ; 17: 1242278, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37901799

RESUMEN

Objective: This study evaluated the antihyperalgesic and anti-inflammatory effects of percutaneous vagus nerve electrical stimulation (pVNS) associated with physical exercise, i.e., swimming, in mice with peripheral inflammation. Methods: The pain model was induced by intraplantar (i.pl.) injection of Freund's complete adjuvant (CFA). Sixty-four male Swiss mice (35-40 g) received an i.pl. of CFA and underwent behavioral tests, i.e., mechanical hyperalgesia, edema, and paw temperature tests. Additionally, cytokine levels, specifically interleukin-6 (IL-6) and interleukin-10 (IL-10), were determined by enzyme-linked immunosorbent assay. Mice were treated with swimming exercise for 30 min alone or associated with different time protocols (10, 20, or 30 min) of stimulation in the left ear with random frequency during four consecutive days. Results: pVNS for 20 min prolonged the antihyperalgesic effect for up to 2 h, 24 h after CFA injection. pVNS for 30 min prolonged the antihyperalgesic effect for up to 7 h, 96 h after CFA injection. However, it did not alter the edema or temperature at both analyzed times (24 and 96 h). Furthermore, the combination of pVNS plus swimming exercise, but not swimming exercise alone, reduced IL-6 levels in the paw and spinal cord, as well as IL-10 levels in the spinal cord. Conclusion: pVNS potentiates the analgesic effect induced by swimming, which may be, at least in part, mediated by the modulation of inflammatory cytokines in the periphery (paw) and central nervous system (spinal cord). Therefore, the combination of these therapies may serve as an important adjunctive treatment for persistent inflammatory pain.

2.
ACS Chem Neurosci ; 14(10): 1859-1869, 2023 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-37116219

RESUMEN

Cav3.2 channels play an important role in the afferent nociceptive pathway, which is responsible for both physiological and pathological pain transmission. Cav3.2 channels are upregulated during neuropathic pain or peripheral inflammation in part due to an increased association with the deubiquitinase USP5. In this study, we investigated nine naturally occurring flavonoid derivatives which we tested for their abilities to inhibit transiently expressed Cav3.2 channels and their interactions with USP5. Icariside II (ICA-II), one of the flavonols studied, inhibited the biochemical interactions between USP5 and Cav3.2 and concomitantly and effectively blocked Cav3.2 channels. Molecular docking analysis predicts that ICA-II binds to the cUBP domain and the Cav3.2 interaction region. In addition, ICA-II was predicted to interact with residues in close proximity to the Cav3.2 channel's fenestrations, thus accounting for the observed blocking activity. In mice with inflammatory and neuropathic pain, ICA-II inhibited both phases of the formalin-induced nocifensive responses and abolished thermal hyperalgesia induced by injection of complete Freund's adjuvant (CFA) into the hind paw. Furthermore, ICA-II produced significant and long-lasting thermal anti-hyperalgesia in female mice, whereas Cav3.2 null mice were resistant to the action of ICA-II. Altogether, our data show that ICA-II has analgesic activity via an action on Cav3.2 channels.


Asunto(s)
Canales de Calcio Tipo T , Neuralgia , Femenino , Ratones , Animales , Canales de Calcio Tipo T/metabolismo , Simulación del Acoplamiento Molecular , Neuralgia/tratamiento farmacológico , Neuralgia/metabolismo , Hiperalgesia/metabolismo , Flavonoides , Flavonoles , Ratones Noqueados , Proteasas Ubiquitina-Específicas/metabolismo
3.
ACS Chem Neurosci ; 13(4): 524-536, 2022 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-35113527

RESUMEN

Cav3.2 calcium channels are important mediators of nociceptive signaling in the primary afferent pain pathway, and their expression is increased in various rodent models of chronic pain. Previous work from our laboratory has shown that this is in part mediated by an aberrant expression of deubiquitinase USP5, which associates with these channels and increases their stability. Here, we report on a novel bioactive rhodanine compound (II-1), which was identified in compound library screens. II-1 inhibits biochemical interactions between USP5 and the Cav3.2 domain III-IV linker in a dose-dependent manner, without affecting the enzymatic activity of USP5. Molecular docking analysis reveals two potential binding pockets at the USP5-Cav3.2 interface that are distinct from the binding site of the deubiquitinase inhibitor WP1130 (a.k.a. degrasyn). With an understanding of the ability of some rhodanines to produce false positives in high-throughput screening, we have conducted several orthogonal assays to confirm the validity of this hit, including in vivo experiments. Intrathecal delivery of II-1 inhibited both phases of formalin-induced nocifensive behaviors in mice, as well as abolished thermal hyperalgesia induced by the delivery of complete Freund's adjuvant (CFA) to the hind paw. The latter effects were abolished in Cav3.2 null mice, thus confirming that Cav3.2 is required for the action of II-1. II-1 also mediated a robust inhibition of mechanical allodynia induced by injury to the sciatic nerve. Altogether, our data uncover a novel class of analgesics─well suited to rapid structure-activity relationship studies─that target the Cav3.2/USP5 interface.


Asunto(s)
Analgésicos , Canales de Calcio Tipo T , Neuralgia , Proteasas Ubiquitina-Específicas , Analgésicos/farmacología , Animales , Bloqueadores de los Canales de Calcio , Canales de Calcio Tipo T/metabolismo , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Ratones , Simulación del Acoplamiento Molecular , Neuralgia/metabolismo , Relación Estructura-Actividad , Proteasas Ubiquitina-Específicas/antagonistas & inhibidores , Proteasas Ubiquitina-Específicas/metabolismo
4.
Mol Brain ; 14(1): 166, 2021 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-34775970

RESUMEN

T-type calcium channels are known molecular targets of certain phytocannabinoids and endocannabinoids. Here we explored the modulation of Cav3.2 T-type calcium channels by terpenes derived from cannabis plants. A screen of eight commercially available terpenes revealed that camphene and alpha-bisabolol mediated partial, but significant inhibition of Cav3.2 channels expressed in tsA-201 cells, as well as native T-type channels in mouse dorsal root ganglion neurons. Both compounds inhibited peak current amplitude with IC50s in the low micromolar range, and mediated an additional small hyperpolarizing shift in half-inactivation voltage. When delivered intrathecally, both terpenes inhibited nocifensive responses in mice that had received an intraplantar injection of formalin, with alpha-bisabolol showing greater efficacy. Both terpenes reduced thermal hyperalgesia in mice injected with Complete Freund's adjuvant. This effect was independent of sex, and absent in Cav3.2 null mice, indicating that these compounds mediate their analgesic properties by acting on Cav3.2 channels. Both compounds also inhibited mechanical hypersensitivity in a mouse model of neuropathic pain. Hence, camphene and alpha-bisabolol have a wide spectrum of analgesic action by virtue of inhibiting Cav3.2 T-type calcium channels.


Asunto(s)
Canales de Calcio Tipo T , Neuralgia , Animales , Monoterpenos Bicíclicos/farmacología , Hiperalgesia , Ratones , Sesquiterpenos Monocíclicos , Neuralgia/tratamiento farmacológico , Terpenos/farmacología , Terpenos/uso terapéutico
5.
Mol Brain ; 11(1): 60, 2018 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-30340616

RESUMEN

Cav3.2 T-type calcium channels are important for the signaling of nociceptive information in the primary afferent pain pathway. During neuropathy and peripheral inflammation, Cav3.2 channels are upregulated due to an increased association with the deubiquitinase USP5. Disrupting these interactions in male mice by the use of cell permeant peptides reverses mechanical and thermal hypersensitivity. Here we explore the effects of interfering with USP5 binding to the channel in female mice with synchronized estrous cycle. We show that intrathecal delivery of a cell-penetrating TAT peptide corresponding to the UBPc domain of USP5 fully reverses mechanical hypersensitivity in mice intraplantarly injected with Complete Freund's Adjuvant. Hence, the USP5 mediated dysregulation of Cav3.2 channel activity does not exhibit sex differences, and potential therapeutics targeting this interaction should be effective in both male and female subjects.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Hiperalgesia/metabolismo , Inflamación/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Animales , Dolor Crónico/metabolismo , Dolor Crónico/patología , Femenino , Hiperalgesia/patología , Inflamación/patología , Masculino , Ratones Endogámicos C57BL , Péptidos/metabolismo
6.
Mol Pain ; 122016.
Artículo en Inglés | MEDLINE | ID: mdl-27130589

RESUMEN

BACKGROUND: Cav3.2 T-type calcium currents in primary afferents are enhanced in various painful pathological conditions, whereas inhibiting Cav3.2 activity or expression offers a strategy for combating the development of pain hypersensitivity. We have shown that Cav3.2 channel surface density is strongly regulated by the ubiquitination machinery and we identified the deubiquitinase USP5 as a Cav3.2 channel interacting protein and regulator of its cell surface expression. We also reported that USP5 is upregulated in chronic pain conditions. Conversely, preventing its binding to the channel in vivo mediates analgesia in inflammatory and neuropathic pain models. RESULTS: To identify which USP5 domain is responsible for the interaction, we used a series of USP5-derived peptides corresponding to different regions in nUBP, cUBP, UBA1, and UBA2 domains to outcompete full length USP5. We identified a stretch of amino acid residues within the cUBP domain of USP5 as responsible for binding to Cav3.2 calcium channels. Based on this information, we generated a TAT-cUBP1-USP5 peptide that could disrupt the Cav3.2/USP5 interaction in vitro and tested its physiological effect in well-established models of persistent inflammatory pain (CFA test) and chronic mononeuropathy and polyneuropathy in mice (partial sciatic nerve injury and the (ob/ob) diabetic spontaneous neuropathic mice). Our results reveal that the TAT-cUBP1-USP5 peptide attenuated mechanical hyperalgesia induced by both Complete Freund's Adjuvant and partial sciatic nerve injury, and thermal hyperalgesia in diabetic neuropathic animals. In contrast, Cav3.2 null mice were not affected by the peptide in the partial sciatic nerve injury model. Cav3.2 calcium channel levels in diabetic mice were reduced following the administration of the TAT-cUBP1-USP5 peptide. CONCLUSIONS: Our findings reveal a crucial region in the cUBP domain of USP5 that is important for substrate recognition and binding to the III-IV linker of Cav3.2 channels. Targeting the interaction of this region with the Cav3.2 channel can be exploited as the basis for therapeutic intervention into inflammatory and neuropathic pain.


Asunto(s)
Péptidos de Penetración Celular/uso terapéutico , Endopeptidasas/química , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Neuralgia/complicaciones , Neuralgia/tratamiento farmacológico , Secuencia de Aminoácidos , Animales , Canales de Calcio Tipo T/metabolismo , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/farmacología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/patología , Modelos Animales de Enfermedad , Hiperalgesia/complicaciones , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/patología , Inflamación/patología , Masculino , Ratones Endogámicos C57BL , Neuralgia/patología , Unión Proteica , Dominios Proteicos , Mapeo de Interacción de Proteínas , Nervio Ciático/efectos de los fármacos , Nervio Ciático/lesiones , Nervio Ciático/patología
7.
Mol Pain ; 122016.
Artículo en Inglés | MEDLINE | ID: mdl-27053601

RESUMEN

BACKGROUND: T-type calcium channels are important contributors to signaling in the primary afferent pain pathway and are thus important targets for the development of analgesics. It has been previously reported that certain piperazine-based compounds such as flunarizine are able to inhibit T-type calcium channels. Thus, we hypothesized that novel piperazine compounds could potentially act as analgesics. RESULTS: Here, we have created a series of 14 compound derivatives around a diphenyl methyl-piperazine core pharmacophore. Testing their effects on transiently expressed Cav3.2 calcium channels revealed one derivative (3-((4-(bis(4-fluorophenyl)methyl)piperazin-1-yl)methyl)-4-(2-methoxyphenyl)-1,2,5-oxadiazole 2-oxide, compound 10e) as a potent blocker. 10e mediate tonic block of these channels with an IC50 of around 4 micromolar. 10e also blocked Cav3.1 and Cav3.3 channels, but only weakly affected high-voltage-activated Cav1.2 and Cav2.2 channels. Intrathecal delivery of 10e mediated relief from formalin and complete Freund's adjuvant induced inflammatory pain that was ablated by genetic knockout of Cav3.2 channels. CONCLUSIONS: Altogether, our data identify a novel T-type calcium channel blocker with tight structure activity relationship (SAR) and relevant in vivo efficacy in inflammatory pain conditions.


Asunto(s)
Analgésicos/síntesis química , Analgésicos/farmacología , Bloqueadores de los Canales de Calcio/síntesis química , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo T/metabolismo , Piperazinas/síntesis química , Piperazinas/farmacología , Dolor Agudo/tratamiento farmacológico , Dolor Agudo/fisiopatología , Analgésicos/uso terapéutico , Animales , Bloqueadores de los Canales de Calcio/uso terapéutico , Fenómenos Electrofisiológicos/efectos de los fármacos , Células HEK293 , Humanos , Inyecciones Espinales , Masculino , Ratones Endogámicos C57BL , Piperazinas/uso terapéutico , Factores de Tiempo
8.
Mol Pain ; 10: 77, 2014 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-25481027

RESUMEN

BACKGROUND: T-type calcium channels and cannabinoid receptors are known to play important roles in chronic pain, making them attractive therapeutic targets. We recently reported on the design, synthesis and analgesic properties of a novel T-type channel inhibitor (NMP-7), which also shows mixed agonist activity on CB1 and CB2 receptors in vitro. Here, we analyzed the analgesic effect of systemically delivered NMP-7 (intraperitoneal (i.p.) or intragstric (i.g.) routes) on mechanical hypersensitivity in inflammatory pain induced by Complete Freund's Adjuvant (CFA) and neuropathic pain induced by sciatic nerve injury. RESULTS: NMP-7 delivered by either i.p. or i.g. routes produced dose-dependent inhibition of mechanical hyperalgesia in mouse models of inflammatory and neuropathic pain, without altering spontaneous locomotor activity in the open-field test at the highest active dose. Neither i.p. nor i.g. treatment reduced peripheral inflammation per se, as evaluated by examining paw edema and myeloperoxidase activity. The antinociception produced by NMP-7 in the CFA test was completely abolished in CaV3.2-null mice, confirming CaV3.2 as a key target. The analgesic action of intraperitoneally delivered NMP-7 was not affected by pretreatment of mice with the CB1 antagonist AM281, but was significantly attenuated by pretreatment with the CB2 antagonist AM630, suggesting that CB2 receptors, but not CB1 receptors are involved in the action of NMP-7 in vivo. CONCLUSIONS: Overall, our work shows that NMP-7 mediates a significant analgesic effect in a model of persistent inflammatory and chronic neuropathic pain by way of T-type channel modulation and CB2 receptor activation. Thus, this study provides a novel therapeutic avenue for managing chronic pain conditions via mixed CB ligands/T-type channel blockers.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Carbazoles/química , Inflamación/metabolismo , Neuralgia/tratamiento farmacológico , Receptor Cannabinoide CB2/metabolismo , Analgesia/métodos , Analgésicos/química , Animales , Peso Corporal , Carbazoles/farmacología , Adyuvante de Freund/metabolismo , Hiperalgesia , Inflamación/tratamiento farmacológico , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuralgia/metabolismo , Dolor/tratamiento farmacológico , Dimensión del Dolor , Nervio Ciático/lesiones
9.
Neuron ; 83(5): 1144-58, 2014 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-25189210

RESUMEN

T-type calcium channels are essential contributors to the transmission of nociceptive signals in the primary afferent pain pathway. Here, we show that T-type calcium channels are ubiquitinated by WWP1, a plasma-membrane-associated ubiquitin ligase that binds to the intracellular domain III-IV linker region of the Cav3.2 T-type channel and modifies specific lysine residues in this region. A proteomic screen identified the deubiquitinating enzyme USP5 as a Cav3.2 III-IV linker interacting partner. Knockdown of USP5 via shRNA increases Cav3.2 ubiquitination, decreases Cav3.2 protein levels, and reduces Cav3.2 whole-cell currents. In vivo knockdown of USP5 or uncoupling USP5 from native Cav3.2 channels via intrathecal delivery of Tat peptides mediates analgesia in both inflammatory and neuropathic mouse models of mechanical hypersensitivity. Altogether, our experiments reveal a cell signaling pathway that regulates T-type channel activity and their role in nociceptive signaling.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Endopeptidasas/metabolismo , Inflamación/fisiopatología , Neuralgia/enzimología , Animales , Canales de Calcio Tipo T/genética , Células Cultivadas , Modelos Animales de Enfermedad , Endopeptidasas/genética , Adyuvante de Freund/toxicidad , Humanos , Hiperalgesia/diagnóstico , Hiperalgesia/fisiopatología , Técnicas In Vitro , Inflamación/inducido químicamente , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Neuralgia/tratamiento farmacológico , Umbral del Dolor/efectos de los fármacos , Umbral del Dolor/fisiología , Péptidos/uso terapéutico , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/fisiología , Médula Espinal/citología , Transfección , Ubiquitinación/genética , Ubiquitinación/fisiología
10.
Pain Med ; 13(8): 1049-58, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22776137

RESUMEN

OBJECTIVE: Investigate whether ankle joint mobilization (AJM) decreases hypersensitivity in the mouse plantar incision (PI) model of postoperative pain as well as to analyze the possible mechanisms involved in this effect. DESIGN: Experiment 1: PI male Swiss mice (25-35 g, N = eight animals per group) were subjected to five sessions of AJM, each lasting either 9 or 3 minutes. AJM movement was applied at a grade III as defined by Maitland. Paw withdrawal frequency to mechanical stimuli was assessed before realization of PI and before and after daily AJM sessions. Mechanical hypersensitivity was also assessed following systemic (intraperitoneal [i.p.]) and local (intraplantar) injection of naloxone (a nonselective opioid receptor antagonist; 1 mg/kg, i.p.; 5 µg/paw, respectively, experiment 2); and systemic injection of fucoidin (100 µg/mouse, i.p., an inhibitor of leukocyte rolling, experiment 3) in different groups of mice. RESULTS: Nine but not 3 minutes of AJM reduced mechanical hypersensitivity caused by PI, an effect that was prevented by systemic and local administrations of naloxone but not by fucoidin. CONCLUSIONS: Our results indicate that joint mobilization reduces postoperative pain by activation of the peripheral opioid pathway. However, antihypersensitivity induced by AJM is apparently not limited by the number of opioid-containing leukocytes but by opioid receptors availability in sensory neurons. A better understanding of the peripheral mechanisms of AJM could stimulate therapists to integrate joint mobilization with strategies also known to influence endogenous pain control, such as exercise, acupuncture, and transcutaneous electrical nerve stimulation to potentiate endogenous analgesia.


Asunto(s)
Articulación del Tobillo/inervación , Hiperalgesia/terapia , Manipulaciones Musculoesqueléticas/métodos , Dolor Postoperatorio/terapia , Receptores Opioides/fisiología , Animales , Articulación del Tobillo/fisiología , Modelos Animales de Enfermedad , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/rehabilitación , Masculino , Ratones , Dolor Postoperatorio/tratamiento farmacológico , Dolor Postoperatorio/rehabilitación
11.
Basic Clin Pharmacol Toxicol ; 104(4): 306-15, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19281602

RESUMEN

The present study examined the antinociceptive effects of a hydroalcoholic extract of Polygala paniculata in chemical and thermal behavioural models of pain in mice. The antinociceptive effects of hydroalcoholic extract was evaluated in chemical (acetic-acid, formalin, capsaicin, cinnamaldehyde and glutamate tests) and thermal (tail-flick and hot-plate test) models of pain or by biting behaviour following intratecal administration of both ionotropic and metabotropic agonists of excitatory amino acids receptors glutamate and cytokines such as interleukin-1beta (IL-1beta) and tumour necrosis factor-alpha (TNF-alpha) in mice. When given orally, hydroalcoholic extract (0.001-10 mg/kg), produced potent and dose-dependent inhibition of acetic acid-induced visceral pain. In the formalin test, the hydroalcoholic extract (0.0001-0.1 mg/kg orally) also caused significant inhibition of both the early (neurogenic pain) and the late (inflammatory pain) phases of formalin-induced licking. However, it was more potent and efficacious in relation to the late phase of the formalin test. The capsaicin-induced nociception was also reduced at a dose of only 1.0 mg/kg orally. The hydroalcoholic extract significantly reduced the cinnamaldehyde-induced nociception at doses of 0.01, 0.1 and 1.0 mg/kg orally. Moreover, the hydroalcoholic extract (0.001-1.0 mg/kg orally) caused significant and dose-dependent inhibition of glutamate-induced pain. However, only rutin, but not phebalosin or aurapten, isolated from P. paniculata, administered intraperitoneally to mice, produced dose-related inhibition of glutamate-induced pain. Furthermore, the hydroalcoholic extract (0.1-100 mg/kg orally) had no effect in the tail-flick test. On the other hand, the hydroalcoholic extract caused a significant increase in the latency to response at a dose of 10 mg/kg orally, in the hot-plate test. The hydroalcoholic extract (0.1 mg/kg orally) antinociception, in the glutamate test, was neither affected by intraperitoenal treatment of animals with l-arginine (precursor of nitric oxide, 600 mg/kg) and naloxone (opioid receptor antagonist, 1 mg/kg) nor associated with non-specific effects such as muscle relaxation or sedation. In addition, oral administration of hydroalcoholic extract produced a great inhibition of the pain-related behaviours induced by intrathecal injection of glutamate, N-methyl-D-aspartate (NMDA), IL-1beta and TNF-alpha, but not by alpha-amino-3-hydroxy-5-methyl-isoxazole-4-propionic acid (AMPA), kainate or trans-1-amino-1.3-cyclopentanediocarboxylic acid (trans-ACPD). Together, our results suggest that inhibition of glutamatergic ionotropic receptors, may account for the antinociceptive action reported for the hydroalcoholic extract from P. paniculata in models of chemical pain used in this study.


Asunto(s)
Analgésicos/farmacología , Dolor/tratamiento farmacológico , Extractos Vegetales/farmacología , Polygala/química , Administración Oral , Analgésicos/administración & dosificación , Analgésicos/aislamiento & purificación , Animales , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inyecciones Intraperitoneales , Masculino , Ratones , Dimensión del Dolor , Extractos Vegetales/administración & dosificación , Receptores de Glutamato/efectos de los fármacos , Receptores de Glutamato/metabolismo , Rutina/administración & dosificación , Rutina/aislamiento & purificación , Rutina/farmacología
12.
Basic Clin Pharmacol Toxicol ; 101(2): 127-31, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17651315

RESUMEN

Cipura paludosa (Iridaceae), a native plant widely distributed in the north of Brazil, is used in traditional medicine as an anti-inflammatory and analgesic agent, against tuberculosis and gonorrhoea and for regulation of menstrual flow. However, scientific studies on the pharmacological properties of C. paludosa are scarce. We have examined the potential protective effects of the ethanolic extract of C. paludosa against methyl mercury (MeHg)-induced neurotoxicity in adult mice. MeHg was diluted in drinking water (40 mg/l, freely available) and the ethanolic C. paludosa extract (CE) was diluted in a 150 mM NaCl solution and administered by gavage (10 and 100 mg/kg body weight, respectively, twice a day). Because treatment lasted for 14 days and each animal weighed around 40 g, the total dosage of plant extract given to each mouse was 5.6 and 56 g, respectively. After the treatment period, MeHg exposure induced a significant deficit in the motor coordination, which was evident by a reduction (90%) in the falling latency in the rotarod apparatus. Interestingly, this phenomenon was completely recovered to control levels by CE co-administration, independent of dosages. MeHg exposure inhibited cerebellar glutathione peroxidase (mean percentage inhibition of 42%) - an important enzyme involved in the detoxification of endogenous peroxides - and this effect was prevented by co-administration of CE. Conversely, MeHg exposure increased cerebellar glutathione reductase activity (mean percentage inhibition of 70%), and this phenomenon was not affected by C. paludosa co-administration. Neither MeHg nor CE changed the cerebellar glutathione levels. This study has shown for the first time, the in vivo protective effects of CE against MeHg-induced neurotoxicity. In addition, our findings encourage studies concerning the beneficial effects of C. paludosa on neurological conditions related to excitotoxicity and oxidative stress.


Asunto(s)
Cerebelo/efectos de los fármacos , Iridaceae , Compuestos de Metilmercurio/antagonistas & inhibidores , Actividad Motora/efectos de los fármacos , Síndromes de Neurotoxicidad/prevención & control , Fitoterapia , Preparaciones de Plantas/uso terapéutico , Animales , Cerebelo/enzimología , Glutatión Peroxidasa/metabolismo , Glutatión Reductasa/metabolismo , Masculino , Compuestos de Metilmercurio/envenenamiento , Ratones , Síndromes de Neurotoxicidad/etiología
13.
J Ethnopharmacol ; 112(1): 19-25, 2007 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-17350194

RESUMEN

This study examined the antinociceptive and anti-inflammatory actions of Cipura paludosa Aubl. in several models of inflammatory pain in mice and rats. The ethanolic extract (EE) from Cipura paludosa (1-300mg/kg) given by i.p. and p.o. routes, 30 or 60min earlier, produced a dose-dependent inhibition of the acetic acid-induced pain and Evans blue leakage in mice with ID(50) values of 2.8 and 17.6mg/kg and 17.2 and 176.1mg/kg, respectively. The EE (10mg/kg, i.p.) also inhibited the allodynia (39+/-6%)- and oedema (97+/-6%)-induced by the intraplantar injection of CFA. In addition, the EE (1-30mg/kg, i.p.) inhibited both mechanical and thermal hyperalgesia induced by prostaglandin E(2), PMA and bradykinin in the rat paw, with ID(50) values of 7.3, 12.1 and 4.7 and 13.9, 18.9 and 1.5mg/kg, respectively. These data demonstrate that EE of Cipura paludosa elicited pronounced antinociceptive and anti-inflammatory actions against some models of inflammatory pain in mice and rats. The mechanism by which the extract produced antinociception still remains unclear, but a great part of this effect seems to be related to modulation of the release or action of pro-inflammatory mediators. Moreover, the antinociceptive action demonstrated in the present study supports, at least partly, the ethnomedical uses of this plant.


Asunto(s)
Analgésicos/farmacología , Antiinflamatorios/farmacología , Iridaceae , Dolor/tratamiento farmacológico , Analgésicos/uso terapéutico , Animales , Antiinflamatorios/uso terapéutico , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Ratones , Dolor/etiología , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Ratas , Ratas Wistar
14.
Brain Res ; 1093(1): 116-22, 2006 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-16765330

RESUMEN

This study was designed to evaluate the role of spinal glutamatergic receptors in the antinociception elicited by agmatine in mice. Intraperitoneal (i.p.) administration of agmatine (1.0-100.0 mg/kg) dose dependently inhibited the nociceptive response induced by intrathecal (i.t.) injection of glutamate, N-methy-D-aspartate (NMDA) and (+/-)-1-aminocyclopentane-trans-1,3-dicarboxylic acid (trans-ACPD), with mean ID(50) values of 16.7, 6.8 and 27.0 mg/kg, respectively. However, agmatine completely failed to affect the nociception induced by alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) or kainic acid (kainate). Agmatine injected by i.t. route (10-100 microg/site) also produced dose-related inhibition of NMDA- and trans-ACPD-induced biting response with mean ID(50) values of 29.6 and 36.0 mug/site, respectively. The nitric oxide synthase inhibitor N(omega)-nitro-L-arginine (l-NOARG) (75.0 mg/kg, i.p.) also consistently inhibited glutamate-, NMDA- and trans-ACPD-induced nociception (41 +/- 13, 100 and 83 +/- 6%, of inhibition, respectively) but had no effect on the same response caused by AMPA and kainate agonists. The selective NMDA receptor antagonist (5S,10R)-(+)-5-Methyl-10,11-dihydro-5H-dibenzo[a,d] (MK-801) at a low dose (0.05 mg/kg, i.p.) inhibited the nociceptive response caused by both glutamate and NMDA agonists (inhibitions of 35 +/- 1 and 72 +/- 2%, respectively). At a high dose, MK-801 (0.5 mg/kg, i.p.) significantly inhibited the biting response induced by i.t. administration of all the glutamatergic agonists tested: glutamate, AMPA, NMDA, kainate and trans-ACPD, with inhibitions of 49 +/- 8, 84 +/- 16, 84 +/- 3, 76 +/- 8 and 97 +/- 2%, respectively. Together, these results provide experimental evidence indicating that agmatine given systemically and spinally produce marked antinociception at spinal sites in mice. Furthermore, an interaction with glutamate receptors, namely NMDA and trans-ACPD, metabotropic and NMDA-ionotropic origin, by a mechanism similar to that of nitric oxide (NO) inhibitors, seems to account for the agmatine antinociceptive action.


Asunto(s)
Agmatina/farmacología , Analgésicos/farmacología , Dolor/fisiopatología , Receptores de Glutamato/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Animales , Cicloleucina/análogos & derivados , Cicloleucina/farmacología , Relación Dosis-Respuesta a Droga , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Femenino , Ácido Glutámico/farmacología , Ácido Kaínico/farmacología , Masculino , Ratones , N-Metilaspartato/farmacología , Fármacos Neuroprotectores/farmacología , Nociceptores/efectos de los fármacos , Nociceptores/metabolismo , Receptores de Glutamato/metabolismo , Médula Espinal/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/farmacología
15.
J Pharm Pharmacol ; 57(10): 1345-51, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16259764

RESUMEN

This study examined the antinociceptive effect of Bauhinia microstachya (Leguminosae), a native plant widely distributed in the South of Brazil, in several chemical and mechanical models of pain. The methanolic extract (ME) from B. microstachya (3--30 mg kg(-1), i.p.) and the isolated compound quercitrin (1--10 mg kg(-1), i.p.), given 30 min earlier, produced a dose-dependent inhibition of acetic-acid-induced visceral pain in mice, with a mean ID50 value (dose necessary to reduce the nociceptive response by 50% relative to the control value) of 7.9 and 2.4 mg kg(-1), respectively. The ME of B. microstachya (3--100 mg kg(-1), i.p., 30 min earlier) also caused a dose-dependent inhibition of capsaicin-induced pain, with a mean ID50 value of 18.8 mg kg(-1). Moreover, the ME (3--100 mg kg(-1), i.p., 30 min earlier) produced marked inhibition of both phases of formalin-induced pain, with mean ID50 values for the neurogenic and the inflammatory phases of 30.3 and 17.2 mg kg(-1), respectively. In addition, the ME of B. microstachya (3--300 mg kg(-1), i.p., 30 min earlier) inhibited, in a graded manner, the hyperalgesia induced by bradykinin (3.2 microg/paw), substance P (13.5 microg/paw), carrageenan (300 microg/paw), capsaicin (100 microg/paw) and adrenaline (100 ng/paw) in the rat paw, with mean ID50 values of 20.5, 17.9, 101.8, 54.2 and 99.7 mg kg(-1), respectively. Taken together, these data demonstrate that ME of B. microstachya elicited a pronounced antinociceptive action against several chemical and mechanical models of pain in mice and rats. The precise mechanism responsible for the antinociceptive effect of the extract still remains unclear, but seems to be partly related to modulation of the release or action of pro-inflammatory mediators involved in the models of pain used. Finally, the flavonoid quercitrin isolated from this plant appears to contribute for the antinociceptive property of the methanolic extract.


Asunto(s)
Bauhinia/química , Extractos Vegetales/farmacología , Hojas de la Planta/química , Quercetina/análogos & derivados , Músculos Abdominales/efectos de los fármacos , Músculos Abdominales/fisiopatología , Ácido Acético , Analgésicos/química , Analgésicos/aislamiento & purificación , Analgésicos/farmacología , Animales , Carragenina , Constricción Patológica/inducido químicamente , Constricción Patológica/fisiopatología , Relación Dosis-Respuesta a Droga , Formaldehído , Hiperalgesia/inducido químicamente , Hiperalgesia/prevención & control , Inyecciones Intraperitoneales , Ratones , Dolor/inducido químicamente , Dolor/prevención & control , Dimensión del Dolor/métodos , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación , Quercetina/química , Quercetina/aislamiento & purificación , Quercetina/farmacología , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA