Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Nutrients ; 13(6)2021 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-34072678

RESUMEN

The incidence of metabolic and chronic diseases including cancer, obesity, inflammation-related diseases sharply increased in the 21st century. Major underlying causes for these diseases are inflammation and oxidative stress. Accordingly, natural products and their bioactive components are obvious therapeutic agents for these diseases, given their antioxidant and anti-inflammatory properties. Research in this area has been significantly expanded to include chemical identification of these compounds using advanced analytical techniques, determining their mechanism of action, food fortification and supplement development, and enhancing their bioavailability and bioactivity using nanotechnology. These timely topics were discussed at the 20th Frontier Scientists Workshop sponsored by the Korean Academy of Science and Technology, held at the University of Hawaii at Manoa on 23 November 2019. Scientists from South Korea and the U.S. shared their recent research under the overarching theme of Bioactive Compounds, Nanoparticles, and Disease Prevention. This review summarizes presentations at the workshop to provide current knowledge of the role of natural products in the prevention and treatment of metabolic diseases.


Asunto(s)
Antiinflamatorios , Antioxidantes , Productos Biológicos , Enfermedades Metabólicas , Animales , Suplementos Dietéticos , Humanos , Enfermedades Metabólicas/tratamiento farmacológico , Enfermedades Metabólicas/metabolismo , Ratones , Nanopartículas , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas
2.
J Cell Physiol ; 233(9): 6984-6995, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29345310

RESUMEN

The use of supplements, such as porcine follicular fluid (pFF), fetal bovine serum and human serum albumin are widely used during in vitro maturation (IVM) in different species but these supplements contain undefined components that cause technical difficulties in standardization and influence the efficiency of IVM. Knockout serum replacement (KSR) is a synthetic protein source, without any undefined growth factors or differentiation-promoting factors. Therefore, it is feasible to use KSR as a defined component for avoiding effects of unknown molecules in an IVM system. In this study, the rates of oocyte maturation and blastocyst formation after parthenogenetic activation (PA), somatic cell nuclear transfer (SCNT) and in vitro fertilization (IVF) were significantly higher in the 5% KSR supplemented group than in the unsupplemented control group and more similar to those of the 10% pFF supplemented group. Moreover, the intensity of GDF9, BMP15, ROS, GSH, BODIPY-LD, BODIPY-FA, and BODIPY-ATP staining showed similar values between 5% KSR and 10% pFF, which have significant difference with control group. Most of the gene expression related to lipid metabolism with both supplements exhibited similar patterns. In conclusion, 5% KSR upregulated lipid metabolism and thereby provides an essential energy source to sustain and improve oocyte quality and subsequent embryo development after PA, SCNT, and IVF. These indications support the idea that KSR used as a defined serum supplement for oocyte IVM might be universally used in other species.


Asunto(s)
Líquido Folicular/metabolismo , Técnicas de Maduración In Vitro de los Oocitos , Metabolismo de los Lípidos , Suero/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Proteína Morfogenética Ósea 15/metabolismo , Compuestos de Boro/metabolismo , Proliferación Celular , Células del Cúmulo/citología , Células del Cúmulo/metabolismo , Desarrollo Embrionario , Femenino , Fertilización In Vitro , Fluorescencia , Regulación de la Expresión Génica , Glutatión/metabolismo , Factor 9 de Diferenciación de Crecimiento/metabolismo , Metabolismo de los Lípidos/genética , Técnicas de Transferencia Nuclear , Oocitos/citología , Oocitos/metabolismo , Partenogénesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Porcinos
3.
Int J Cardiol ; 198: 187-95, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26163916

RESUMEN

BACKGROUND: Mesenchymal stem cells (MSCs) have the potential to differentiate into multiple cell lineages. Given this potential for tissue regeneration, MSC-based therapeutic applications have been considered in recent years. However, ischemia-induced apoptosis has been reported to be one of the main causes of MSC death following transplantation. The primary objective of this study was to determine whether a natural antioxidant, fucoidan, could protect MSCs from ischemia-induced apoptosis in vitro and in vivo. Furthermore, we investigated the mechanism of action of fucoidan's anti-ischemic effect in MSCs. METHODS AND RESULT: Pre-treatment with fucoidan (10 µg/mL) suppressed the increase in H2O2-induced reactive oxygen species (ROS) levels and drastically reduced apoptotic cell death in MSCs. Fucoidan inhibited the activation of the pro-apoptotic proteins p38-mitogen-activated protein kinase (MAPK), Jun N-terminal kinase (JNK), and caspase-3, and augmented the expression of the anti-apoptosis protein cellular inhibitor of apoptosis (cIAP). Moreover, fucoidan significantly increased manganese superoxide dismutase (MnSOD) expression and decreased cellular ROS levels via the Akt pathway, resulting in enhanced cell survival. In a murine hindlimb ischemia model, transplanted fucoidan-treated MSCs showed significantly enhanced cell survival and proliferation in ischemic tissues. Functional recovery and limb salvage also remarkably improved in mice injected with fucoidan-stimulated MSCs compared with mice injected with non-stimulated MSCs. CONCLUSION: Taken together, these results show that fucoidan protects MSCs from ischemia-induced cell death by modulation of apoptosis-associated proteins and cellular ROS levels through regulation of the MnSOD and Akt pathways, suggesting that fucoidan could be powerful therapeutic adjuvant for MSC-based therapy in ischemic diseases.


Asunto(s)
Miembro Posterior/irrigación sanguínea , Isquemia/tratamiento farmacológico , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Polisacáridos/uso terapéutico , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Relación Dosis-Respuesta a Droga , Humanos , Isquemia/patología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/fisiología , Estrés Oxidativo/fisiología , Polisacáridos/farmacología
4.
Artículo en Inglés | MEDLINE | ID: mdl-24089621

RESUMEN

The injection of diluted bee venom (DBV) into an acupoint has been used traditionally in eastern medicine to treat a variety of inflammatory chronic pain conditions. We have previously shown that DBV had a potent antinociceptive efficacy in several rodent pain models. However, the peripheral mechanisms underlying DBV-induced antinociception remain unclear. The present study was designed to investigate the role of peripheral epinephrine on the DBV-induced antinociceptive effect in the mouse formalin assay. Adrenalectomy significantly enhanced the antinociceptive effect of DBV during the late phase of the formalin test, while chemical sympathectomy had no effect. Intraperitoneal injection of epinephrine blocked this adrenalectomy-induced enhancement of the DBV-induced antinociceptive effect. Moreover, injection of a phenylethanolamine N-methyltransferase (PNMT) inhibitor enhanced the DBV-induced antinociceptive effect. Administration of nonselective ß -adrenergic antagonists also significantly potentiated this DBV-induced antinociception, in a manner similar to adrenalectomy. These results demonstrate that the antinociceptive effect of DBV treatment can be significantly enhanced by modulation of adrenal medulla-derived epinephrine and this effect is mediated by peripheral ß -adrenoceptors. Thus, DBV acupoint stimulation in combination with inhibition of peripheral ß -adrenoceptors could be a potentially novel strategy for the management of inflammatory pain.

5.
J Cell Physiol ; 223(2): 397-407, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20112290

RESUMEN

The role of individual supplements necessary for the long-term self-renewal of embryonic stem (ES) cells is poorly characterized in feeder/serum-free culture systems. This study sought to characterize the relationship between the effects of glucose on ES cell proliferation and fibronectin (FN) synthesis, and to assess the mechanisms responsible for these cellular effects of glucose. Treatment of the two ES cells (ES-E14TG2a and ES-R1) with 25 mM glucose (high glucose) increased the expression levels of FN mRNA and protein. In addition, high glucose and ANG II synergistically increased FN expression level, which coincident with data showing that high glucose increased the mRNA expression of angiotensin II (ANG II) type 1 receptor (AT(1)R), angiotensinogen, and FN, but not ANG II type 2 receptor. High glucose also increased the intracellular calcium (Ca(2+)) concentration and pan-protein kinase C (PKC) phosphorylation. Inhibition of the Ca(2+)/PKC pathway blocked high glucose-induced FN expression. High glucose or ANG II also synergistically increased transforming growth factor-beta1 (TGF-beta(1)) expression, while pretreatment with losartan abolished the high glucose-induced increase in TGF-beta(1) production. Moreover, TGF-beta(1)-specific small interfering RNA inhibited high glucose-induced FN expression and c-Jun N-terminal kinase (JNK) activation. The JNK inhibitor SP600125 blocked high glucose-induced FN expression and inhibited cell cycle regulatory protein expression induced by high glucose or TGF-beta(1). In this study, inhibition of AT(1)R, Ca(2+)/PKC, TGF-beta(1), JNK, FN receptor blocked the high glucose-induced DNA synthesis, increased the cell population in S phase, and the number of cells. It is concluded that high glucose increases FN synthesis through the ANG II or TGF-beta1 pathways, which in part mediates proliferation of mouse ES cells.


Asunto(s)
Angiotensina II/metabolismo , Proliferación Celular/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Fibronectinas/biosíntesis , Glucosa/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Angiotensina II/farmacología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Angiotensinógeno/genética , Animales , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Recuento de Células , Replicación del ADN/efectos de los fármacos , Replicación del ADN/genética , Células Madre Embrionarias/efectos de los fármacos , Fibronectinas/efectos de los fármacos , Fibronectinas/genética , Glucosa/farmacología , Hiperglucemia/genética , Hiperglucemia/metabolismo , Hiperglucemia/fisiopatología , Proteínas Quinasas JNK Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Losartán/farmacología , Ratones , Fosforilación/efectos de los fármacos , Proteína Quinasa C/efectos de los fármacos , Proteína Quinasa C/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Receptor de Angiotensina Tipo 1/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta1/efectos de los fármacos , Factor de Crecimiento Transformador beta1/genética
6.
J Pain ; 10(3): 253-63, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19010737

RESUMEN

UNLABELLED: Although intrathecal (i.t.) administration of the alpha(2)-adrenoceptor agonist clonidine has a pronounced analgesic effect, the clinical use of clonidine is limited by its side effects. Previously, our laboratory has demonstrated that the subcutaneous injection of diluted bee venom (DBV) into an acupoint (termed apipuncture) produces significant analgesic effect in various pain animal models. The present study was designed to examine whether DBV injection into the Zusanli acupoint (ST-36) could enhance lower-dose clonidine-induced analgesic effects without the development of hypotension, bradycardia, or sedation. In the mouse formalin test, DBV injection produced a dramatic leftward shift in the dose-response curve for clonidine-induced analgesia. In a rat neuropathic pain model i.t. clonidine dose dependently suppressed chronic constriction injury (CCI)-induced mechanical allodynia and thermal hyperalgesia, and this clonidine-induced analgesic effect was significantly potentiated by apipuncture pretreatment. DBV apipuncture alone or in combination with a low dose of i.t. clonidine produced an analgesic effect similar to that of the high dose of clonidine, but without significant side effects. The analgesic effect produced by the combination of i.t. clonidine and apipuncture was completely blocked by pretreatment with an alpha(2)-adrenoceptor antagonist. These data show that DBV-apipuncture significantly enhances clonidine-induced analgesia and suggest that a combination of low dose clonidine with acupuncture therapy represents a novel strategy for pain management that could eliminates clonidine's side effects. PERSPECTIVE: This study demonstrated that intrathecal clonidine-induced analgesia is significantly enhanced when it is combined with chemical acupuncture treatment. The administration of low-dose clonidine in combination with acupuncture produced a potent analgesic effect without significant side effects and thus represents a potential novel strategy for the management of chronic pain.


Asunto(s)
Puntos de Acupuntura , Analgésicos/uso terapéutico , Venenos de Abeja/uso terapéutico , Clonidina/uso terapéutico , Hiperalgesia/tratamiento farmacológico , Dimensión del Dolor , Dolor/tratamiento farmacológico , Analgésicos/efectos adversos , Animales , Venenos de Abeja/administración & dosificación , Clonidina/administración & dosificación , Clonidina/efectos adversos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Hiperalgesia/inducido químicamente , Inyecciones Espinales , Inyecciones Subcutáneas , Masculino , Ratones , Ratones Endogámicos ICR , Dolor/inducido químicamente , Dolor/fisiopatología , Dimensión del Dolor/métodos , Ratas , Ratas Sprague-Dawley
7.
J Pharm Pharmacol ; 60(10): 1347-54, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18812028

RESUMEN

Ursolic acid (UA) is pentacyclic triterpenoic acid that naturally occurs in many medicinal herbs and plants. In this study, we examined the possible suppressive effect of UA extracted from Oldenlandia diffusa on zymosan-induced acute inflammation in mice and complete Freund's adjuvant (CFA)-induced arthritis in rats. UA treatment (per oral) dose-dependently (25-200 mg kg(-1)) suppressed zymosan-induced leucocyte migration and prostaglandin E2 (PGE(2)) production in the air pouch exudates. Since the maximal effective dose of UA was 50 mg kg(-1) in the zymosan experiment, we used this dose of UA in a subsequent study using an adjuvant-induced rheumatoid arthritis model. UA treatment (50 mg kg(-1), per oral, once a day for 10 days) was started from day 12 after adjuvant injection. UA dramatically inhibited paw swelling, plasma PGE(2) production and radiological changes in the joint caused by CFA injection. Moreover, UA significantly suppressed the arthritis-induced mechanical and thermal hyperalgesia as well as the spinal Fos expression, as determined by immunohistochemistry, which was increased by CFA injection. In addition, overall anti-arthritic potency of UA was comparable with ibuprofen (100 mg kg(-1), oral) while UA did not induce significant gastric lesions as compared with the ibuprofen treatment group. These findings strongly suggest that UA is a useful suppressive compound for rheumatoid arthritis treatment with low risk of gastric problems.


Asunto(s)
Antirreumáticos/uso terapéutico , Artritis Experimental/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Triterpenos/uso terapéutico , Enfermedad Aguda , Animales , Antirreumáticos/química , Artritis Experimental/fisiopatología , Movimiento Celular/efectos de los fármacos , Enfermedad Crónica , Dinoprostona/biosíntesis , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inflamación/inducido químicamente , Inflamación/fisiopatología , Leucocitos/citología , Leucocitos/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Oldenlandia/química , Dolor/fisiopatología , Dolor/prevención & control , Extractos Vegetales/química , Extractos Vegetales/uso terapéutico , Ratas , Ratas Sprague-Dawley , Triterpenos/química , Zimosan , Ácido Ursólico
8.
Brain Res Bull ; 75(5): 698-705, 2008 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-18355649

RESUMEN

Although the frequency-dependent antinociceptive mechanisms of electroacupuncture (EA) have been well demonstrated, the anti-inflammatory mechanisms that underlie the suppressive effects induced by different frequencies of EA stimulation on peripheral inflammation are largely unknown. We have previously reported that EA stimulation can activate the sympathetic nervous system (SNS) and that this activation is responsible for the EA-induced suppression of zymosan-induced leukocyte migration. The present study was designed to evaluate the differential effect of low (1Hz, LF EA) versus high (120Hz, HF EA) frequency EA stimulation on SNS activation and ultimately on carrageenan-induced inflammation. Immediately after carrageenan injection, we applied either LF EA or HF EA bilaterally to the Zusanli (ST36) acupoints. To evaluate the anti-inflammatory effect of EA (EA-AI), paw volume and myeloperoxidase (MPO) activity, a marker of infiltrated leukocytes, were measured and the paw withdrawal latency to noxious heat stimulation was also assessed. Both LF EA and HF EA significantly suppressed the carrageenan-induced paw edema and MPO activity. Moreover, thermal hyperalgesia was strongly attenuated in both the LF EA and HF EA groups. Adrenalectomy significantly diminished HF EA-AI without affecting LF EA-AI. Pretreatment with the corticosterone receptor antagonist, RU-486 did not affect either LF EA- or HF EA-AI. On the other hand, administration of 6-hydroxydopamine (a neurotoxin for peripheral sympathetic nerve endings) selectively blocked LF EA-AI. Propranolol (a beta-adrenoceptor antagonist) completely abolished both LF EA- and HF EA-AI. The results of this study suggest that the suppressive effects of LF EA on carrageenan-induced paw inflammation are mediated by sympathetic post-ganglionic neurons, while the suppressive effects of HF EA are mediated by the sympatho-adrenal medullary axis.


Asunto(s)
Médula Suprarrenal/fisiología , Estimulación Eléctrica/métodos , Electroacupuntura/métodos , Ganglios Simpáticos/patología , Inflamación/patología , Inflamación/terapia , Neuronas/efectos de la radiación , Médula Suprarrenal/efectos de la radiación , Adrenalectomía , Adrenérgicos/administración & dosificación , Animales , Carragenina , Modelos Animales de Enfermedad , Relación Dosis-Respuesta en la Radiación , Edema/inducido químicamente , Edema/patología , Antagonistas de Hormonas/administración & dosificación , Inflamación/inducido químicamente , Masculino , Ratones , Ratones Endogámicos ICR , Mifepristona/administración & dosificación , Neuronas/fisiología , Umbral del Dolor/efectos de los fármacos , Umbral del Dolor/fisiología , Umbral del Dolor/efectos de la radiación , Peroxidasa/metabolismo , Factores de Tiempo
9.
Neurosci Lett ; 430(2): 163-8, 2008 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-18061346

RESUMEN

Our recent data, obtained using a zymosan-induced inflammatory air pouch model in mice, have demonstrated that subcutaneous bee venom (BV) injection into the hind limb selectively activates the contralateral brain stem locus coeruleus (LC) and then via a descending noradrenergic pathway and subsequent adrenal medullary catecholamine release induces a potent anti-inflammatory effect. While the efferent limb of this BV-induced neuroimmune anti-inflammatory pathway is well documented, the afferent limb of this pathway is poorly understood. In particular the spinal mechanisms involved with BV activation of the LC are currently unknown. Spinal nitric oxide (NO) and its synthase (NOS) have been shown to play an important role in the transmission and amplification of neuronal information from the spinal cord to the brain stem. In the present study we evaluated whether spinal NO plays a role in BV-induced LC activation, since we have previously shown that LC activation underlies this 'BV-induced anti-inflammatory effect' (BVAI) using the mouse air pouch model. Intrathecal (i.t.) pretreatment with l-nitro arginine methyl ester (l-NAME, non-selective NOS inhibitor), hemoglobin (NO scavenger) or 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ, soluble guanylate cyclase inhibitor) abolished BVAI on zymosan-induced leukocyte migration into the air pouch. Moreover, i.t. injection of l-N-iminoethyl-lysine (l-NIL, inducible NOS inhibitor), but not 7-nitroindazole (7-NI, neuronal NOS inhibitor), also inhibited BVAI. BV injection significantly increased both the number of Fos immunoreactive neurons and tyrosine hydroxylase-Fos double labeling neurons in the contralateral LC in zymosan-induced inflamed mice. Importantly this increase in Fos expression in the LC was also completely inhibited by i.t. injection of l-NIL, but not by i.t. injection of 7-NI. Collectively these results indicate that spinal NO generated from inducible NOS is involved in the BV-induced LC activation that underlies BVAI.


Asunto(s)
Antiinflamatorios/uso terapéutico , Venenos de Abeja/uso terapéutico , Locus Coeruleus/enzimología , Óxido Nítrico/metabolismo , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Médula Espinal/enzimología , Animales , Modelos Animales de Enfermedad , Interacciones Farmacológicas , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Indazoles/farmacología , Leucocitos/efectos de los fármacos , Locus Coeruleus/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos ICR , NG-Nitroarginina Metil Éster/farmacología , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/enzimología , Proteínas Oncogénicas v-fos/metabolismo , Médula Espinal/efectos de los fármacos , Tirosina 3-Monooxigenasa/metabolismo
10.
Brain Res ; 1148: 69-75, 2007 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-17367766

RESUMEN

Electroacupuncture (EA) is used to treat a variety of inflammatory diseases; however, the neurophysiological mechanisms underlying EA's anti-inflammatory effect remain unclear. Accumulating evidence suggests that the sympathetic nervous system regulates immunologic and inflammatory responses and thus we hypothesized that this system could be involved in EA's anti-inflammatory effect (EA-AI). The goal of the present study was to evaluate whether the sympathetic nervous system plays a critical role in EA-AI using a mouse air pouch inflammation model. We found that bilateral low-frequency (1 Hz) EA applied to the Zusanli acupoint significantly suppressed the number of zymosan-induced leukocytes migrating into the air pouch. Furthermore, double-labeling immunohistochemical experiments showed that EA stimulation increased Fos expression in choline acetyltransferase (ChAT)-positive sympathetic pre-ganglionic neurons in the intermediolateral region of thoracic spinal cord segments. Chemical sympathetic denervation by intraperitoneal injection of 6-hydroxydopamine (which spares sympathetic adrenal medullary innervation) significantly inhibited EA-AI. In contrast, adrenalectomy did not alter EA-AI. Finally, systemic propranolol administration significantly inhibited EA's anti-inflammatory effect, suggesting that beta-adrenoceptors are involved. Collectively, these results suggest that EA produces an anti-inflammatory effect in this mouse air pouch model by activating the sympathetic nervous system leading to the release of catecholamines from post-ganglionic nerve terminals, which act on beta-adrenoceptors on immune cells to inhibit their migration.


Asunto(s)
Electroacupuntura/métodos , Inflamación/fisiopatología , Inflamación/terapia , Piel/inervación , Piel/fisiopatología , Fibras Simpáticas Posganglionares/metabolismo , Acetilcolinesterasa/metabolismo , Antagonistas Adrenérgicos beta/farmacología , Animales , Catecolaminas/metabolismo , Quimiotaxis de Leucocito/efectos de los fármacos , Quimiotaxis de Leucocito/fisiología , Modelos Animales de Enfermedad , Electroacupuntura/normas , Inflamación/inducido químicamente , Mediadores de Inflamación/efectos adversos , Masculino , Ratones , Ratones Endogámicos ICR , Neuronas/metabolismo , Terminales Presinápticos/metabolismo , Piel/efectos de los fármacos , Médula Espinal/metabolismo , Simpatectomía Química/métodos , Zimosan/efectos adversos
11.
J Pain ; 7(7): 500-12, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16814689

RESUMEN

UNLABELLED: Peripheral bee venom (BV) administration produces 2 contrasting effects, nociception and antinociception. This study was designed to evaluate whether the initial nociceptive effect induced by BV injection into the Zusanli acupoint is involved in producing the more prolonged antinociceptive effect observed in the mouse formalin test, and whether capsaicin-sensitive primary afferents are involved in these effects. BV injection into the Zusanli point increased spinal Fos expression but not spontaneous nociceptive behavior. BV pretreatment 10 minutes before intraplantar formalin injection dose-dependently attenuated nociceptive behavior associated with the second phase of the formalin test. The destruction of capsaicin-sensitive primary afferents by resiniferatoxin (RTX) pretreatment selectively decreased BV-induced spinal Fos expression but did not affect BV-induced antinociception. Furthermore, BV injection increased Fos expression in tyrosine hydroxylase immunoreactive neurons in the locus caeruleus, and this expression was unaltered by RTX pretreatment. Finally, BV's antinociception was blocked by intrathecal injection of 10 microg idazoxan, and this effect was not modified by RTX pretreatment. These findings suggest that subcutaneous BV stimulation of the Zusanli point activates central catecholaminergic neurons via capsaicin-insensitive afferent fibers without induction of nociceptive behavior. This in turn leads to the activation of spinal alpha2-adrenoceptors, which ultimately reduces formalin-evoked nociceptive behaviors. PERSPECTIVE: This study demonstrates that BV acupuncture produces a significant antinociception without nociceptive behavior in rodents, which is mediated by capsaicin-insensitive afferents and involves activation of central adrenergic circuits. These results further suggest that BV stimulation into this acupuncture point might be a valuable alternative to traditional electrical or mechanical acupoint stimulation.


Asunto(s)
Analgesia por Acupuntura/métodos , Vías Aferentes/efectos de los fármacos , Venenos de Abeja/farmacología , Neuronas Aferentes/efectos de los fármacos , Nociceptores/efectos de los fármacos , Dolor/tratamiento farmacológico , Analgesia por Acupuntura/normas , Puntos de Acupuntura , Vías Aferentes/fisiología , Analgésicos/farmacología , Analgésicos/uso terapéutico , Animales , Venenos de Abeja/uso terapéutico , Capsaicina/farmacología , Modelos Animales de Enfermedad , Diterpenos/farmacología , Resistencia a Medicamentos/efectos de los fármacos , Resistencia a Medicamentos/fisiología , Locus Coeruleus/efectos de los fármacos , Locus Coeruleus/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Neuronas Aferentes/fisiología , Neurotoxinas/farmacología , Nociceptores/fisiología , Norepinefrina/biosíntesis , Dolor/fisiopatología , Dimensión del Dolor/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores Adrenérgicos alfa 2/efectos de los fármacos , Receptores Adrenérgicos alfa 2/metabolismo , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
12.
Neurosci Res ; 55(2): 197-203, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16621078

RESUMEN

Several lines of evidence indicate significant interactions between the immune and nervous systems. Our recent study reveals that 'bee venom (BV) induced anti-inflammatory effect' (BVAI) was produced by sympathetic preganglionic neuronal activation and subsequent adrenomedullary catecholamine release in a zymosan-induced inflammation model. However, the specific peripheral input and the supraspinal neuronal systems that are involved in this BVAI remain to be defined. Here we show that subcutaneous BV injection into left hind limb significantly reduces zymosan-induced leukocyte migration and that this effect is completely inhibited by denervation of the left sciatic nerve. This BVAI was not affected by the destruction of capsaicin-sensitive primary afferent fibers using either neonatal capsaicin or resiniferatoxin (RTX) pretreatment. BV injection into the left hind limb significantly increased Fos expression in the contralateral locus coeruleus (LC) in non-inflamed mice. In zymosan-inflamed mice, BV injection produced a further increase in LC Fos expression as compared with non-inflamed mice. This BV-induced Fos increase in the LC was not affected by RTX pretreatment. Pharmacological blockage of central noradrenergic activity by either central chemical sympathectomy (i.c.v. 6-hydroxydopamine) or alpha2 adrenoceptor antagonism (i.c.v. idazoxan) completely blocked BVAI. Taken together, these results suggest that BVAI is mediated by peripheral activation of capsaicin-insensitive primary afferent fibers and subsequent central noradrenergic activation including the LC.


Asunto(s)
Antiinflamatorios/uso terapéutico , Venenos de Abeja/uso terapéutico , Inflamación/tratamiento farmacológico , Locus Coeruleus/efectos de los fármacos , Norepinefrina/metabolismo , Adrenérgicos/farmacología , Animales , Capsaicina/farmacología , Modelos Animales de Enfermedad , Diterpenos/farmacología , Interacciones Farmacológicas , Lateralidad Funcional , Expresión Génica/efectos de los fármacos , Inflamación/inducido químicamente , Leucocitos/efectos de los fármacos , Leucocitos/fisiología , Locus Coeruleus/metabolismo , Ratones , Ratones Endogámicos ICR , Proteínas Oncogénicas v-fos/metabolismo , Oxidopamina/farmacología , Neuropatía Ciática/dietoterapia , Neuropatía Ciática/patología , Neuropatía Ciática/fisiopatología , Zimosan
13.
J Altern Complement Med ; 12(1): 39-44, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16494567

RESUMEN

BACKGROUND: Although acupuncture has been widely used for complementary therapeutic approaches to treat inflammatory diseases and inflammation-induced pain, the potential anti-inflammatory effects of acupuncture treatment remain controversial in clinical trials, and the underlying mechanisms are still unclear. OBJECTIVE: The objective was to determine whether electroacupuncture (EA) is able to suppress the peripheral inflammatory response (e.g., zymosan-induced leukocyte migration into air pouch). As part of a mechanistic approach, it was further evaluated whether endogenous opioid systems are involved in the "EA-induced anti-inflammatory effect" (EA-AI). METHODS: EA (1 or 120 Hz) was performed bilaterally in the Zusanli acupoint (ST36) or in a nonacupoint (gluteal muscle) for 30 min in ICR mice under anesthetic condition. The number of leukocytes that migrated into the air pouch was counted 4 hours after zymosan injection. EA was performed at 0, 0.5, 1, or 2 hours prior to zymosan injection, respectively. To evaluate opioid involvement in EA-AI, intrathecal naloxone (36 microg/mouse) and intraperitoneal naloxone methiodide (30 mg/kg) were administered 10 min before EA stimulation. RESULTS: Both the 1 and 120 Hz frequencies of EA into Zusanli acupoint at the same time with zymosan injection significantly reduced leukocyte migration into the air pouch as compared with those of control groups (i.e., anesthetic control and needling control into Zusanli acupoint without electrical stimulation). The EA stimulation into nonacupoint did not produce any significant anti-inflammatory effect. EA treatment at 0.5 hours prior to zymosan injection also produced an anti-inflammatory effect but 1 and 2 hours prior to zymosan injection did not elicit any effect. Peripheral opioid blockage significantly reversed EA-AI, whereas spinal opioid blockage did not alter EA-AI. CONCLUSION: EA can suppress peripheral inflammation through a peripheral opioid mechanism. To achieve the full effectiveness of EA, repeated application is recommended for the treatment of a variety of inflammatory diseases.


Asunto(s)
Electroacupuntura/métodos , Inflamación/terapia , Dolor/prevención & control , Receptores Opioides/efectos de la radiación , Análisis de Varianza , Animales , Regulación hacia Abajo , Edema/inducido químicamente , Inflamación/inducido químicamente , Masculino , Ratones , Ratones Endogámicos ICR , Dolor/etiología , Receptores Opioides/metabolismo , Zimosan/toxicidad
14.
Pharmacol Biochem Behav ; 80(1): 181-7, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15652394

RESUMEN

We have previously shown that subcutaneous bee venom (BV) injection reduces visceral pain behavior in mice, but it is not clear which constituent of BV is responsible for its antinociceptive effect. In the present study, we now demonstrate that a water-soluble subfraction of BV (BVA) reproduces the antinociceptive effect of BV in acetic acid-induced visceral pain model. We further evaluated three different BVA subfractions that were separated by molecular weight, and found that only the BVAF3 subfraction (a molecular weight of <10 kDa) produced a significant antinociceptive effect on abdominal stretches and suppressed visceral pain-induced spinal cord Fos expression. Injection of melittin (MEL), a major constituent of BVAF3, also produced a visceral antinociception. However, melittin's antinociception was completely blocked by boiling for 10 min at 100 degrees C, while boiling either whole BV or BVAF3 did not prevent their antinociception. The antinociceptive effect of BVAF3 was completely blocked by intrathecal pretreatment with the alpha2-adrenoceptor antagonist, yohimbine (YOH), while intrathecal pretreatment with the opioid antagonist, naloxone (NAL) or the serotonin antagonist, methysergide, had no effect. These data demonstrate that BVAF3 is responsible for the visceral antinociception of whole BV and further suggest that this effect is mediated in part by spinal alpha2-adrenergic activity.


Asunto(s)
Venenos de Abeja/uso terapéutico , Dolor/tratamiento farmacológico , Receptores Adrenérgicos alfa 2/metabolismo , Médula Espinal/efectos de los fármacos , Animales , Venenos de Abeja/aislamiento & purificación , Venenos de Abeja/farmacología , Abejas , Fraccionamiento Químico , Masculino , Ratones , Ratones Endogámicos ICR , Peso Molecular , Dolor/metabolismo , Solubilidad , Médula Espinal/metabolismo , Vísceras/efectos de los fármacos , Vísceras/fisiología , Agua
15.
Pharmacol Res ; 51(2): 183-8, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15629266

RESUMEN

In a previous report, subcutaneous injection of diluted bee venom (dBV) into a specific acupuncture point (Zusanli, ST36), a procedure termed apipuncture, was shown to produce an antinociceptive effect in the rat formalin pain model. However, the central antinociceptive mechanisms responsible for this effect have not been established. Traditional acupuncture-induced antinociception is considered to be mediated by activation of the descending pain inhibitory system (DPIS) including initiation of its opioidergic, adrenergic and serotonergic components. The purpose of the present study was to investigate whether the antinociceptive effect of apipuncture is also mediated by the DPIS. Behavioral experiments verified that apipuncture significantly reduces licking behavior in the late phase of formalin test in rats. This antinociceptive effect of apipuncture was not modified by intrathecal pretreatment with naltrexone (a non-selective opioid receptor antagonist), prazosin (an alpha1 adrenoceptor antagonist) or propranolol (an beta adrenoceptor antagonist). In contrast, intrathecally injected idazoxan (an alpha2 adrenoceptor antagonist) or intrathecal methysergide (a serotonin receptor antagonist) significantly reversed apipuncture-induced antinociception. These results suggest that apipuncture-induced antinociception is produced by activation of alpha2 adrenergic and serotonergic components of the DPIS.


Asunto(s)
Acupuntura/métodos , Fibras Adrenérgicas/efectos de los fármacos , Analgésicos/farmacología , Venenos de Abeja/farmacología , Dimensión del Dolor/efectos de los fármacos , Serotonina/fisiología , Fibras Adrenérgicas/fisiología , Animales , Abejas , Masculino , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/fisiología , Dimensión del Dolor/métodos , Ratas , Ratas Sprague-Dawley
16.
J Pain ; 5(6): 297-303, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15336634

RESUMEN

UNLABELLED: Chemical acupuncture with diluted bee venom (DBV), termed apipuncture, has been traditionally used in oriental medicine to treat several inflammatory diseases and chronic pain conditions. In the present study we investigated the potential antihyperalgesic and antiallodynic effects of apipuncture in a rat neuropathic pain model. DBV (0.25 mg/kg, subcutaneous) was injected into the Zusanli acupoint 2 weeks after chronic constrictive injury (CCI) of the sciatic nerve. Between 5 and 45 minutes after DBV injection, we observed a significant reduction in the thermal hyperalgesia induced by CCI, but apipuncture failed to reduce CCI-induced mechanical allodynia. We subsequently examined whether this antihyperalgesic effect of apipuncture was related to the activation of spinal opioid receptors and/or alpha2-adrenoceptors. Intrathecal pretreatment with naloxone (10 microg/rat), an opioid receptor antagonist, did not reverse the antihyperalgesic effect of apipuncture, whereas pretreatment with idazoxan (40 microg/rat), an alpha2-adrenoceptor antagonist, completely blocked the effect of apipuncture. These results indicate that DBV-induced apipuncture significantly reduces the thermal hyperalgesia generated by CCI and also suggest that this antihyperalgesic effect is dependent on the activation of alpha2-adrenoceptors, but not opioid receptors, in the spinal cord. PERSPECTIVE: The antinociceptive effect of apipuncture was evaluated in a rodent neuropathic pain model. The relieving effect of apipuncture on thermal hyperalgesia was found to be mediated by spinal alpha2-adrenoceptors, but not opioid receptors. These data suggest that apipuncture might be an effective alternative therapy for patients with painful peripheral neuropathy, especially for those who are poorly responsive to opioid analgesics.


Asunto(s)
Puntos de Acupuntura , Venenos de Abeja/administración & dosificación , Hiperalgesia/terapia , Manejo del Dolor , Receptores Adrenérgicos alfa 2/fisiología , Antagonistas Adrenérgicos/farmacología , Antagonistas de Receptores Adrenérgicos alfa 2 , Animales , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Sprague-Dawley , Médula Espinal/metabolismo
17.
Mol Cells ; 17(2): 329-33, 2004 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-15179050

RESUMEN

Fos immunocytochemistry was combined with tyrosine hydroxylase (TH) or dopamine-beta-hydroxylase (DBH) immunolabeling to examine brainstem catecholaminergic neuronal activation resulting from bee venom (BV) stimulation of the Zusanli acupoint (ST36) in Sprague-Dawley rats. BV injection into the Zusanli acupoint caused increased Fos expression in catecholaminergic neurons located in the hypothalamic arcuate nucleus (Arc), the dorsal raphe (DR), the A5 cell group (A5) and the locus coeruleus (LC). BV acupoint stimulation significantly increased Fos-TH double-labeled neurons in the Arc, LC and DR. Fos-DBH positive neurons were also increased by BV acupoint stimulation in the LC and A5. In contrast BV stimulation of a non-acupoint only increased Fos expression and Fos-TH double-labeled neurons in the Arc. These data indicate that BV acupoint stimulation activates brainstem catecholaminergic neurons and that this activation underlies BV acupoint-induced antinociception.


Asunto(s)
Puntos de Acupuntura , Venenos de Abeja/metabolismo , Tronco Encefálico/metabolismo , Catecolaminas/metabolismo , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Analgésicos/metabolismo , Animales , Tronco Encefálico/citología , Dopamina beta-Hidroxilasa/metabolismo , Inmunohistoquímica , Masculino , Neuronas/citología , Ratas , Ratas Sprague-Dawley , Tirosina 3-Monooxigenasa/metabolismo
18.
J Vet Med Sci ; 65(3): 349-55, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12679565

RESUMEN

In two previous reports, we have demonstrated that injection of bee venom (BV) into an acupoint produces a significant antinociceptive and anti-inflammatory effect in both a mouse model of visceral nociception and a rat model of chronic arthritis. The present study was designed to evaluate the potential antinociceptive effect of BV pretreatment on formalin-induced pain behavior and it associated spinal cord Fos expression in rats. Adult Sprague-Dawley rats were injected with BV directly into the Zusanli (ST36) acupoint or into an arbitrary non-acupoint located on the back. BV pretreatment into the Zusanli acupoint significantly decreased paw-licking time in the late phase of the formalin test. In contrast, BV injected into a non-acupoint in the back region did not suppress the paw-licking time. In addition, BV pretreatment into the Zusanli acupoint markedly inhibited spinal cord Fos expression induced by formalin injection. These findings indicate that BV pretreatment into the Zusanli acupoint has an antinociceptive effect on formalin-induced pain behavior.


Asunto(s)
Puntos de Acupuntura , Venenos de Abeja/farmacología , Formaldehído/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Manejo del Dolor , Dolor/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-fos/metabolismo , Médula Espinal/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Masculino , Dolor/inducido químicamente , Ratas , Ratas Sprague-Dawley
19.
Kidney Blood Press Res ; 25(5): 308-14, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12435877

RESUMEN

Oxidative stress has been implicated as a primary cause of renal failure in certain renal diseases. Indeed, renal proximal tubule is a very sensitive site to oxidative stress and retains functionally fully characterized transporters. It has been reported that ginsenosides have a beneficial effect on diverse diseases including oxidative stress. However, the protective effect of ginsenosides on oxidative stress has not been elucidated in renal proximal tubule cells. Thus, we examined the effect of ginsenosides on oxidative stress-induced alteration of apical transporters and its related mechanism in renal proximal tubule cells. In the present study, hydrogen peroxide (H(2)O(2)) (>10(-5) M) inhibited alpha-methyl-D-glucopyranoside uptake in a dose-dependent manner (p < 0.05). It also inhibited Pi and Na(+) uptake. At a concentration of 20 microg/ml, total ginsenosides significantly reduced H(2)O(2)-induced inhibition of apical transporters. In contrast, protopanaxadiol (PD) and protopanaxatriol (PT) saponins exhibited a less preventive effect than total ginsenosides (p < 0.05). Furthermore, we examined its action mechanism. H(2)O(2) increased lipid peroxide formation, arachidonic acid (AA) release, and Ca(2+) uptake. These effects on H(2)O(2) were significantly prevented by total ginsenosides and PD or PT sanponins. However, total ginsenosides appear to be more protective than PD and PT saponins (p < 0.05). In conclusion, ginsenosides prevented H(2)O(2)-induced inhibition of apical transporters via a decrease in oxidative stress, AA release, and Ca(2+) uptake in primary cultured renal proximal tubule cells.


Asunto(s)
Ginsenósidos/farmacología , Peróxido de Hidrógeno/farmacología , Túbulos Renales Proximales/metabolismo , Proteínas de Transporte de Membrana/efectos de los fármacos , Proteínas de Transporte de Membrana/metabolismo , Animales , Ácido Araquidónico/metabolismo , Calcio/farmacocinética , Relación Dosis-Respuesta a Droga , Peróxido de Hidrógeno/administración & dosificación , Túbulos Renales Proximales/citología , Peróxidos Lipídicos/metabolismo , Masculino , Metilglucósidos/farmacocinética , Estrés Oxidativo/fisiología , Fósforo/farmacocinética , Conejos , Sapogeninas/farmacología , Sodio/farmacocinética , Triterpenos/farmacología
20.
Planta Med ; 68(11): 971-4, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12451485

RESUMEN

Recent epidemiological studies have demonstrated that ginseng intake is associated with a reduced risk for environmentally related cancers. However, the effects of ginsenosides on the proliferation of renal proximal tubule cells have not yet elucidated. This study investigated the effect of total ginsenosides, protopanaxatriol (PT) saponin, and protopanaxadiol (PD) saponin fraction on epidermal growth factor (EGF)-induced renal cell proliferation and, furthermore, c-fos and c-jun gene expression. In the present study, total ginsenosides (10 -6 g/ml) completely blocked EGF-induced DNA synthesis and cell growth. In contrast, the PT and PD fractions partially blocked it. In addition, the EGF-induced increase of c-fos and c-jun gene expression was completely blocked by total ginsenosides and partially by PT and PD saponins. In conclusion, ginsenosides, in part, inhibit EGF-induced cell proliferation via decrease of c-fos and c-jun gene expression in primary cultured rabbit renal proximal tubular cells (PTCs). Abbreviations. EGF:epidermal growth factor PD:protopanaxadiol PT:protopanaxatriol PTCs:primary cultured renal proximal tubule cells


Asunto(s)
División Celular/efectos de los fármacos , Ginsenósidos/farmacología , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/efectos de los fármacos , Panax , Fitoterapia , Sapogeninas/farmacología , Triterpenos/farmacología , Animales , Relación Dosis-Respuesta a Droga , Factor de Crecimiento Epidérmico , Regulación Neoplásica de la Expresión Génica , Genes fos/genética , Genes jun/genética , Ginsenósidos/administración & dosificación , Ginsenósidos/uso terapéutico , Humanos , Neoplasias Renales/prevención & control , Masculino , Raíces de Plantas , Conejos , Sapogeninas/administración & dosificación , Sapogeninas/uso terapéutico , Timidina/metabolismo , Triterpenos/administración & dosificación , Triterpenos/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA