Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Pharmazie ; 74(8): 449-461, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31526436

RESUMEN

Glatiramer acetate is indicated for the treatment of patients with relapsing forms of multiple sclerosis (RMS). In 2016, an alternative to the originator product was approved in the EU through the hybrid procedure regulatory pathway. This paper reviews the scientifically rigorous and multifaceted program undertaken to demonstrate the equivalence of this glatiramer acetate follow-on product (GTR) and the reference product Copaxone®, which resulted in the EU approval of GTR 20 mg/mL and 40 mg/mL. Establishing therapeutic equivalence for non-biological complex drugs is not trivial and requires a complex and multidisciplinary effort. Ultimately, there is not a single test or study that establishes therapeutic equivalence of two heterogeneous products. Instead, it requires a good understanding of the synthesis process together with a full set of data that includes comparative physicochemical testing, nonclinical in vitro and in vivo studies, and a comparative clinical study to allow for a valid conclusion that two products are therapeutically equivalent. The detailed understanding of glatiramer's synthesis process and its impact on the characteristics of glatiramer, combined with the results of a scientifically rigorous and multifaceted physicochemical and biological characterization program, and the clinical data from the 794-patient Phase III GATE study, demonstrate that GTR and Copaxone are therapeutically equivalent. The data further demonstrate that Synthon's manufacturing process consistently yields drug substance of the same quality as Copaxone and that switching from Copaxone to GTR is safe and well-tolerated.


Asunto(s)
Acetato de Glatiramer/farmacología , Inmunosupresores/farmacología , Animales , Ensayos Clínicos Fase III como Asunto , Método Doble Ciego , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/genética , Expresión Génica , Acetato de Glatiramer/farmacocinética , Humanos , Inmunosupresores/farmacocinética , Ratones , Ensayos Clínicos Controlados Aleatorios como Asunto , Ratas , Células THP-1 , Equivalencia Terapéutica
2.
Osteoarthritis Cartilage ; 27(3): 484-492, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30576794

RESUMEN

There is a need for efficacious and safe pain treatments for OA (osteoarthritis). The nerve growth factor (NGF) antibody tanezumab is associated with high efficacy, but when combined with chronic NSAID treatment shows an increased risk of rapidly progressive osteoarthritis (RPOA) in a small group of patients. AIM: The aim of this study was to phenotype with biochemical biomarkers of bone, cartilage, soft tissue, synovial metabolism OA patients who are at risk of developing RPOA type-2, for both limited and chronic NSAIDs users. MATERIAL AND METHODS: The dataset consisted of OA patients participating in tanezumab trials who used NSAIDs <90 days (limited NSAID users) or chronic users (NSAIDs ≥90 days) over an average 10 month period. Biomarker data were available for 47 cases (RPOA type-2) and 92 controls. Non-linear and linear multivariable predictive models were developed. RESULTS: By use of two biomarkers at baseline the receiver operating characteristic (ROC) curve area for RPOA type-2 in limited NSAID users was 71%, [CI] (60-83%). OA subjects with this biomarker phenotype had 8-fold higher confidence interval [CI][(2-33)] relative risk of developing RPOA type-2 as compared to OA patients without this phenotype. The AUC of the model in chronic NSAIDs users based on 5 biomarkers was 78%, [CI](69-88%), with 4-fold [CI (2-6)] relative risk of developing RPOA type-2. CONCLUSION: In this hypothesis generating and exploratory study, we identified combinations of biomarkers associated with OA patients who develop RPOA type-2, which may be related to the pathology of the RPOA type-2 joint.


Asunto(s)
Analgésicos/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Osteoartritis/sangre , Antiinflamatorios no Esteroideos/uso terapéutico , Biomarcadores/sangre , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Persona de Mediana Edad , Osteoartritis/tratamiento farmacológico , Oxicodona/uso terapéutico
3.
Osteoarthritis Cartilage ; 25(6): 866-877, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28115232

RESUMEN

OBJECTIVE: Establish a biomarker panel associated with all-cause total joint replacement (TJR) through identification of patients with osteoarthritis (OA) who do or do not progress to TJR and investigate effects of nonsteroidal anti-inflammatory drugs (NSAIDs). DESIGN: Serum samples from patients enrolled in phase III trials of tanezumab who experienced TJR (n = 174) or matched patients who did not (n = 321) were analyzed for bone, cartilage, soft tissue, and inflammation markers. Classification and Regression Tree (CART) analysis was used to identify biomarker phenotypes associated with TJR. RESULTS: At baseline, biomarker combinations for patients who did not use NSAIDs before starting tanezumab and used NSAIDs during tanezumab treatment <90 days ("nonNSAID"), identified 77% (95% confidence interval [CI]: 71-84%) of patients who experienced TJR and 77% (95% CI: 65-86%) who did not over a 6-month study period (on average). These biomarker combinations increased odds of identifying patients to remain free of a TJR by 3.3-fold. In patients who used NSAIDs continuously (during screening and ≥90 days during tanezumab treatment), 64% (95% CI: 54-73%) who had TJR and 75% (95% CI: 68-83%) who did not were identified by biomarker combinations different from nonNSAID patients, with an increase in odds of identifying patients to remain free of a TJR by two-fold. CONCLUSIONS: Although validation on other cohorts is necessary, biomarkers may assist in identifying patients who will need TJR. The profiles suggest NSAID use increases importance of bone metabolism in TJR pathology.


Asunto(s)
Biomarcadores/sangre , Osteoartritis de la Cadera/sangre , Osteoartritis de la Rodilla/sangre , Proteínas Adaptadoras Transductoras de Señales , Adulto , Anciano , Anciano de 80 o más Años , Analgésicos/uso terapéutico , Antiinflamatorios no Esteroideos/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Artroplastia de Reemplazo de Cadera , Artroplastia de Reemplazo de Rodilla , Proteínas Morfogenéticas Óseas/sangre , Proteína C-Reactiva/metabolismo , Proteína de la Matriz Oligomérica del Cartílago/sangre , Colágeno Tipo I/sangre , Colágeno Tipo II/sangre , Colágeno Tipo III/sangre , Progresión de la Enfermedad , Femenino , Marcadores Genéticos , Humanos , Péptidos y Proteínas de Señalización Intercelular/sangre , Interleucina-6/sangre , Modelos Logísticos , Masculino , Metaloproteinasa 9 de la Matriz/sangre , Persona de Mediana Edad , Osteoartritis de la Cadera/terapia , Osteoartritis de la Rodilla/terapia , Osteocalcina/sangre , Fragmentos de Péptidos/sangre , Péptidos/sangre , Procolágeno/sangre , Pronóstico , Puntaje de Propensión , Factor A de Crecimiento Endotelial Vascular/sangre
4.
Clin Exp Immunol ; 180(1): 28-39, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25393803

RESUMEN

Current therapies for multiple sclerosis (MS), a chronic autoimmune neuroinflammatory disease, mostly target general cell populations or immune molecules, which may lead to a compromised immune system. A more directed strategy would be to re-enforce tolerance of the autoaggressive T cells that drive tissue inflammation and injury. In this study, we have investigated whether the course of experimental autoimmune encephalomyelitis (EAE) in mice and marmosets can be altered by a potent tolerizing fusion protein. In addition, a multi-parameter immunological analysis was performed in marmosets to assess whether the treatment induces modulation of EAE-associated cellular and humoral immune reactions. The fusion protein, CTA1R9K-hMOG10-60-DD, contains a mutated cholera toxin A1 subunit (CTA1R9K), a dimer of the Ig binding D region of Staphylococcus aureus protein A (DD), and the human myelin oligodendrocyte glycoprotein (hMOG) sequence 10-60. We observed that intranasal application of CTA1R9K-hMOG10-60-DD seems to skew the immune response against myelin oligodendrocyte glycoprotein (MOG) towards a regulatory function. We show a reduced number of circulating macrophages, reduced MOG-induced expansion of mononuclear cells in peripheral blood, reduced MOG-induced production of interleukin (IL)-17A in spleen, increased MOG-induced production of IL-4 and IL-10 and an increased percentage of cells expressing programmed cell death-1 (PD-1) and CC chemokine receptor 4 (CCR4). Nevertheless, the treatment did not detectably change the EAE course and pathology. Thus, despite a detectable effect on relevant immune parameters, the fusion protein failed to influence the clinical and pathological outcome of disease. This result warrants further development and improvement of this specifically targeted tolerance inducing therapy.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Inmunomodulación/efectos de los fármacos , Esclerosis Múltiple/tratamiento farmacológico , Glicoproteína Mielina-Oligodendrócito/farmacología , Animales , Callithrix , Toxina del Cólera/genética , Toxina del Cólera/inmunología , Toxina del Cólera/farmacología , Citocinas/genética , Citocinas/inmunología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Inmunomodulación/genética , Ratones , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Glicoproteína Mielina-Oligodendrócito/genética , Glicoproteína Mielina-Oligodendrócito/inmunología , Compuestos Orgánicos , Receptores CCR4/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacología , Proteína Estafilocócica A/genética , Proteína Estafilocócica A/inmunología , Proteína Estafilocócica A/farmacología
5.
J Mol Endocrinol ; 32(3): 689-701, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15171709

RESUMEN

With the aim of developing an in vivo model that directly detects activation of estrogen receptors (ERs), transgenic mice carrying a luciferase reporter gene were generated. The luciferase reporter gene was under the control of three consensus estrogen-responsive elements (EREs) coupled to a minimal TATA-box, with or without flanking chick beta-globin insulators. By using this model in combination with the IVIS imaging system, in vivo ER activation was measured. Dose- and time-dependent luciferase activity was induced in various organs of adult transgenic male mice exposed to diethylstilbestrol (DES) (10-1000 micro g/kg) and 17beta-estradiol dipropionate (EP) (10-1000 micro g/kg), when luciferase activity was measured ex vivo. The highest (>10 000-fold) induction of luciferase was measured in bone and kidney 24 h after exposure to 1000 micro g/kg EP. Other highly responsive organs include liver, testis, pituitary, brain, prostate and colon, which show different activity profiles. This in vivo model for detecting estrogenic activity can be used to assess tissue-specific action of ER agonists and antagonists. These could include selective ER modulators and environmental estrogens. In combination with the IVIS imaging system, this in vivo model is a powerful tool for assessing the kinetics of gene activation by estrogenic compounds.


Asunto(s)
Regulación de la Expresión Génica , Genes Reporteros , Ratones Transgénicos , Receptores de Estrógenos/metabolismo , Animales , Dietilestilbestrol/metabolismo , Estradiol/metabolismo , Estrógenos no Esteroides/metabolismo , Femenino , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Receptores de Estrógenos/genética , Elementos de Respuesta , Factores de Tiempo , Distribución Tisular , Activación Transcripcional
6.
J Endocrinol ; 169(2): 271-80, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11312144

RESUMEN

Carp beta-endorphin is posttranslationally modified by N-terminal acetylation and C-terminal cleavage. These processes determine the biological activity of the beta-endorphins. Forms of beta-endorphin were identified in the pars intermedia and the pars distalis of the pituitary gland of the common carp (Cyprinus carpio), as well as the forms released in vitro and into the blood. After separation and quantitation by high performance liquid chromatography (HPLC) coupled with radioimmunoassay, the beta-endorphin immunoreactive products were identified by electrospray ionisation mass spectrometry and peptide sequencing. The release of beta-endorphins by the pituitary gland was studied after stimulation with corticotrophin-releasing factor (CRF) in vitro. In the pars intermedia, eight N-acetylated truncated forms were identified. Full length N-acetyl beta-endorphin(1-33) coeluted with N-acetyl beta-endorphin(1-29) and these forms together amounted to over 50% of total immunoreactivity. These products were partially processed to N-acetyl betaendorphin(1-15) (30.8% of total immunoreactivity) and N-acetyl beta-endorphin(1-10) (3.1%) via two different cleavage pathways. The acetylated carp homologues of mammalian alpha- and gamma-endorphin were also found. N-acetyl beta-endorphin(1-15) and (1-29) and/or (1-33) were the major products to be released in vitro, and were the only acetylated beta-endorphins found in blood plasma, although never together. CRF stimulated the release of opioid beta-endorphin from the pars distalis. This non-acetylated beta-endorphin represents the full length peptide and is the most abundant form in plasma.


Asunto(s)
Carpas/metabolismo , Hipófisis/química , betaendorfina/análogos & derivados , betaendorfina/análisis , Animales , Cromatografía Líquida de Alta Presión , Hormona Liberadora de Corticotropina/farmacología , Inmunohistoquímica , Técnicas In Vitro , Masculino , Peso Molecular , Hipófisis/anatomía & histología , Hipófisis/metabolismo , Radioinmunoensayo , Espectrometría de Masa por Ionización de Electrospray , Estimulación Química , betaendorfina/sangre
7.
J Endocrinol ; 166(2): 427-35, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10927632

RESUMEN

MSH is a pituitary hormone derived by post-translational processing from POMC and involved in stress and background adaptation. N-terminal acetylation of MSH to monoacetyl alpha-MSH or diacetyl alpha-MSH increases the bioactivity of the peptide. The aim of this study was to characterize alpha-MSH acetylation in the sea bream (Sparus aurata L.) pituitary gland in response to the stressors air exposure and confinement, as well as in fish adapted for 15 days to a white, gray or black background. Pituitary homogenates were purified by reversed-phase HPLC (RP-HPLC). The alpha-MSH content of fractions was measured by RIA. Immunoreactive RP-HPLC fractions were further analyzed by electrospray mass spectrometry and the peptide sequence determined as SYSMEHFRWGKPV-NH2. In the pituitary gland of sea bream, des-, mono- and diacetyl alpha-MSH were identified. Then plasma alpha-MSH levels were measured in sea bream adapted to different backgrounds. Surprisingly, we found the highest plasma alpha-MSH levels in white-adapted as compared with black-adapted sea bream with intermediate values for gray-adapted fish. This observation is in contrast with results that have been obtained in eel, trout or terrestrial vertebrates. Next, des-, mono- and diacetyl alpha-MSH forms were measured in homogenates of the pituitary gland and in plasma of sea bream exposed to air, to confinement, or to different backgrounds. Monoacetyl alpha-MSH was the predominant form in all control and experimental groups. The lowest content of monoacetyl alpha-MSH relative to des- and diacetyl alpha-MSH was found in white-adapted fish. Levels of des- and diacetyl alpha-MSH forms were similar under all conditions. We observed that monoacetyl alpha-MSH is the most abundant isoform in the pituitary gland after background adaptation, confinement and air exposure, in sea bream. These data indicate that the physiologically most potent isoform of alpha-MSH may vary from species to species.


Asunto(s)
Hipófisis/metabolismo , Dorada/metabolismo , alfa-MSH/aislamiento & purificación , Acetilación , Adaptación Fisiológica , Análisis de Varianza , Animales , Cromatografía Líquida de Alta Presión/métodos , Isomerismo , Masculino , Espectrometría de Masas/métodos , Radioinmunoensayo/métodos , Restricción Física , Análisis de Secuencia de Proteína , Estrés Psicológico , alfa-MSH/metabolismo
8.
Anesthesiology ; 92(3): 739-53, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10719953

RESUMEN

BACKGROUND: Despite widespread use, little is known about the comparative pharmacokinetics of intrathecally administered opioids. The present study was designed to characterize the rate and extent of opioid distribution within cerebrospinal fluid, spinal cord, epidural space, and systemic circulation after intrathecal injection. METHODS: Equal doses of morphine and alfentanil, fentanyl, or sufentanil were administered intrathecally (L3) to anesthetized pigs. Microdialysis probes were used to sample cerebrospinal fluid at L2, T11, T7, T3, and the epidural space at L2 every 5-10 min for 4 h. At the end of the experiment, spinal cord and epidural fat tissue were sampled, and each probe's recovery was determined in vitro. Using SAAM II pharmacokinetic modeling software (SAAM Institute, University of Washington, Seattle, WA), the data were fit to a 16-compartment model that was divided into four spinal levels, each of which consisted of a caternary arrangement of four compartments representing the spinal cord, cerebrospinal fluid, epidural space, and epidural fat. RESULTS: Model simulations revealed that the integral exposure (area under the curve divided by dose) of the spinal cord (i.e., effect compartment) to the opioids was highest for morphine because of its low spinal cord distribution volume and slow clearance into plasma The integral exposure of the spinal cord to the other opioids was relatively low, but for different reasons: alfentanil has a high clearance from spinal cord into plasma, fentanyl distributes rapidly into the epidural space and fat, and sufentanil has a high spinal cord volume of distribution. CONCLUSIONS: The four opioids studied demonstrate markedly different pharmacokinetic behavior, which correlates well with their pharmacodynamic behavior.


Asunto(s)
Alfentanilo/farmacocinética , Analgésicos Opioides/farmacocinética , Fentanilo/farmacocinética , Morfina/farmacocinética , Médula Espinal/metabolismo , Sufentanilo/farmacocinética , Alfentanilo/administración & dosificación , Analgésicos Opioides/administración & dosificación , Animales , Fentanilo/administración & dosificación , Inyecciones Espinales , Microdiálisis , Modelos Biológicos , Morfina/administración & dosificación , Sufentanilo/administración & dosificación , Porcinos
9.
J Endocrinol ; 163(1): 149-57, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10495417

RESUMEN

We investigated short-term effects (up to 24 h) of air exposure and confinement, and long-term effects (up to 11 days) of confinement, to elucidate signalling pathways in the stress response of gilthead sea bream Sparus aurata L. Plasma glucose and lactate were taken as indicators of sympathetic activation, and alpha-melanocyte stimulating hormone (alpha-MSH), adrenocorticotrophic hormone (ACTH) and cortisol as indicators of activation of the brain-pituitary-interrenal (BPI) axis. Air exposure for 3 min resulted, within 30 min, in an increase in plasma concentrations of cortisol, alpha-MSH, glucose, lactate, osmolality and plasma Na, Cl and Mg. Plasma ACTH and beta-endorphin and plasma K, Ca and P did not change. We conclude that air exposure mainly activates the brain-sympathetic-chromaffin cell (BSC) axis. In fish confined at a density of 70 kg/m(3) (compared with 4 kg/m(3) in controls), cortisol, ACTH and alpha-MSH increased within 1 h, indicating activation of the BPI axis. Plasma glucose, Na, Cl and Mg increased with an 8 h delay compared with the response to air exposure. No changes in plasma lactate, osmolality, K, Ca and P were observed. Long-term confinement induced a biphasic cortisol response with peaks at 1 h and at 2 and 3 days. A gradual increase in plasma beta-endorphin concentrations peaked at 7 days; the concentration of alpha-MSH increased rapidly within 1 h and then declined to control values 4 days after the onset of confinement. No changes in ACTH were detected. Our data provide evidence that a stressor-specific activation of the BSC and BPI axes may occur in Sparus aurata.


Asunto(s)
Neurotransmisores/sangre , Perciformes/sangre , Transducción de Señal/fisiología , Estrés Fisiológico/fisiopatología , Hormona Adrenocorticotrópica/sangre , Animales , Glucemia/análisis , Hidrocortisona/sangre , Ácido Láctico/análisis , Masculino , Minerales/sangre , Concentración Osmolar , alfa-MSH/análisis , betaendorfina/sangre
10.
Anesthesiology ; 91(1): 97-108, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10422934

RESUMEN

BACKGROUND: The combination of propofol and alfentanil with nitrous oxide provides balanced anesthesia with rapid recovery and minimal emetic side effects. The object of this study was to compare recovery parameters at varying proportions of propofol and alfentanil, and to determine the dosing rate and plasma concentration of propofol necessary to supplement nitrous oxide in the presence of varying concentrations of alfentanil METHODS: Forty-eight patients were anesthetized with nitrous oxide, targeted manual infusions of alfentanil (target plasma concentrations of 0, 50, 100, and 150 ng/ml), and propofol at rates that were varied up or down by 25% depending on the response (movement/no movement) of the preceding patient (at the same alfentanil target concentrations) to ulnar-nerve stimulation. The minimum concentrations of propofol and alfentanil required to prevent movement in 50% of patients (EC50) was determined by logistic regression. Speed of emergence and recovery of cognitive function, time to discharge, and incidence of side effects were compared for four different combinations of propofol and alfentanil with nitrous oxide. RESULTS: The EC50 for propofol alone with nitrous oxide was 6.1 microg/ml. AlfentaniL at concentrations of 41+/-17 (SD), 113+/-54, and 130+/-61 ng/mL reduced the EC50 of propofol to 3.3, 2.3, and 2.2 microg/ml, respectively, and decreased emergence time (eye opening) to 8.1, 4.9, and 3.4 min, compared with 24.3 min for propofol alone. Side effects did not differ between groups. CONCLUSIONS: The authors conclude that there is a synergistic effect between propofol and alfentanil, and that combining alfentanil with propofol is associated with faster early recovery.


Asunto(s)
Alfentanilo/administración & dosificación , Procedimientos Quirúrgicos Ambulatorios , Anestésicos/administración & dosificación , Óxido Nitroso/administración & dosificación , Propofol/administración & dosificación , Adolescente , Adulto , Anciano , Alfentanilo/sangre , Cognición/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Femenino , Humanos , Masculino , Persona de Mediana Edad , Óxido Nitroso/sangre , Propofol/sangre
11.
Mol Cell Endocrinol ; 143(1-2): 23-31, 1998 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-9806347

RESUMEN

Proopiomelanocortin (POMC) is the precursor for a number of biologically active peptides such as adrenocorticotropic hormone (ACTH), alpha-melanocyte-stimulating hormone (alpha-MSH) and beta-endorphin. It is well known that these peptides are involved in the stress response in fish as well as in mammals. We have cloned two different carp POMC cDNAs called, POMC-I and POMC-II. The nucleotide sequences of 955 bp for POMC-I and 959 bp for POMC-II share 93.5% identity in their cDNAs, and the deduced amino acid sequences (both 222 amino acids) are 91.4% identical. In the ACTH and beta-MSH domain, two amino acid substitutions are found, whereas alpha-MSH and beta-endorphin are identical. For beta-MSH, the serine replacement (in POMC-I) by a glycine (in POMC-II) results in a putative amidation site Pro-X-Gly for POMC-II. We used RT-PCR to show that both POMC mRNAs are expressed in the hypophysis, hypothalamus and other parts of the brain of a single fish. Furthermore, in a phylogenetic tree based on POMC sequences the divergence of carp POMC-I and -II from tetraploid animals (salmon, trout and xenopus) is demonstrated.


Asunto(s)
Proopiomelanocortina/genética , ARN Mensajero/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Carpas , Clonación Molecular , ADN Complementario/genética , ADN Complementario/aislamiento & purificación , Datos de Secuencia Molecular , Filogenia , Alineación de Secuencia , Análisis de Secuencia
12.
J Endocrinol ; 159(1): 85-91, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9795345

RESUMEN

Pro-opiomelanocortin (POMC) is the precursor of a number of biologically active peptides, including adrenocorticotropic hormone, alpha-melanocyte-stimulating hormone and beta-endorphin, which are released by the pituitary glands of fish as well as mammals. To quantify the levels of expression of the two POMC mRNAs relative to one another during the response of the common carp to temperature-induced stress, we used reverse transcriptase PCR combined with capillary electrophoresis and laser-induced fluorescence detection. The ratio of POMC-I mRNA to POMC-II mRNA determined in wild-type and four isogenic carp strains was found to be strain-dependent and influenced by temperature. In strain E20xR8, the ratio had altered in favour of POMC-I from 1:3.2 (POMC-I:POMC-II) in fish adapted to 24 degreesC to 1:1.2 in fish adapted to a decrease of 9 degreesC in ambient temperature. A rapid drop in temperature from 24 to 15 degreesC decreased the POMC mRNA ratio at the expense of POMC-I from 1:1.9 in the control fish (strain E4xR3R8) to 1:4.2 3 h after the temperature drop of 9 degreesC. We conclude that both POMC genes are expressed in the common carp and that their expression ratio is strain-dependent and changes in response to ambient temperature.


Asunto(s)
Adaptación Fisiológica , Carpas/metabolismo , Frío , Hipófisis/metabolismo , Proopiomelanocortina/genética , ARN Mensajero/análisis , Animales , Electroforesis Capilar , Femenino , Expresión Génica , Hidrocortisona/análisis , Masculino , Microscopía Fluorescente , Radioinmunoensayo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
J Pharmacol Exp Ther ; 286(2): 585-92, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9694907

RESUMEN

As a follow-up study to an earlier report that racemic fenfluramine can acutely potentiate the analgesic effects of morphine in humans, we investigated the effects of fenfluramine on the development of tolerance to morphine analgesia in rats. Antinociceptive effect, as measured by the tail-flick latency, was studied over 8 days in rats that received continuous i.v. infusion of 1) 22 mg/kg/day of morphine, 2) 20 mg/kg/day of fenfluramine, 3) both drugs concomitantly or 4) saline. Infusion with morphine alone resulted in a peak analgesia of 100% maximal possible effect, which declined with time; full tolerance was reached by day 4. Fenfluramine treatment alone had no effect. Fenfluramine coinfusion attenuated the development of tolerance to morphine; >70% maximal possible effect was still present on day 4. The effect of fenfluramine coinfusion occurred in the absence of a significant increase in plasma or brain morphine concentration, or a decrease in the accumulation of morphine's putative antagonistic metabolite, morphine-3-glucuronide. In another set of infusion experiments, rats were challenged with a single i.p. dose of morphine to characterize the morphine dose-response curves at 10 hr following 4-day i.v. infusion of 1) 22 mg/kg/day of morphine, 2) 20 mg/kg/day fenfluramine, 3) morphine plus fenfluramine or 4) saline. An acute i. p. morphine challenge dose response experiment was also conducted in naïve control rats and in rats receiving a concomitant i.p. injection of fenfluramine (2.4 mg/kg). Coinjection of fenfluramine acutely potentiated the antinociceptive potency of morphine. However, potentiation alone does not fully account for the apparent attenuation of tolerance during morphine i.v. infusion. ED50 of morphine was elevated to 7.0 mg/kg in the morphine-infused rats compared to 2.4 mg/kg in saline-infused rats. Coinfusion of fenfluramine increased ED50 to only 3.7 mg/kg. These results demonstrate that fenfluramine significantly attenuates tolerance development to morphine by modulating the pharmacological process responsible for tolerance development to morphine.


Asunto(s)
Analgésicos Opioides/antagonistas & inhibidores , Fenfluramina/farmacología , Morfina/antagonistas & inhibidores , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Analgésicos Opioides/farmacocinética , Analgésicos Opioides/farmacología , Animales , Cromatografía Líquida de Alta Presión , Relación Dosis-Respuesta a Droga , Tolerancia a Medicamentos , Masculino , Morfina/farmacocinética , Morfina/farmacología , Derivados de la Morfina/farmacología , Dimensión del Dolor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos , Estereoisomerismo , Factores de Tiempo
14.
J Pharmacol Exp Ther ; 279(2): 803-12, 1996 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8930187

RESUMEN

The in vivo concentration-anticonvulsant effect relationships of six benzodiazepines, midazolam, clonazepam, oxazepam, flunitrazepam, diazepam and clobazam were quantified in individual rats and correlated with the affinity to the GABAA-benzodiazepine receptor complex. Furthermore the interaction between midazolam and the benzodiazepine antagonist flumazenil was characterized. All benzodiazepines exhibited a nonlinear relationship between concentration and anticonvulsant effect without ceiling of the effect at higher concentration. The potency of most benzodiazepines was similar with values of the EC250, between 0.067 +/- 0.01 mg. l-1 for midazolam and 0.21 +/- 0.03 mg. l-1 for diazepam. The EC250,u of clobazam was 2.8 +/- 0.9 mg. l-1. These values were considerably larger than the Ki for binding at the GABAA-benzodiazepine receptor complex. No correlation was observed between EC250,u and Ki. Antagonism of the anticonvulsant effect of midazolam by flumazenil was associated with a remarkable change in the concentration-anticonvulsant effect relationship. Analysis of these data on basis of a composite model provided evidence for two separate effects of which only one is antagonized by flumazenil. The anticonvulsant effect at low midazolam concentration was characterized on basis of the sigmoid E maximal effect pharmacodynamic model. The value of the EC50,u was 0.0086 +/- 0.0013 mg. l-1 which is similar to the Ki for binding at the GABAA-benzodiazepine receptor complex. The second more pronounced anticonvulsant effect occurred at higher concentration and was described by an exponential function. The findings of this study indicate that the effect of benzodiazepines against seizures induced by cortical stimulation in vivo cannot be fully accounted for by an interaction at the GABAA-benzodiazepine receptor complex.


Asunto(s)
Anticonvulsivantes/farmacología , Benzodiazepinas/farmacología , Corteza Cerebral/efectos de los fármacos , Animales , Anticonvulsivantes/farmacocinética , Benzodiazepinas/farmacocinética , Corteza Cerebral/fisiología , Relación Dosis-Respuesta a Droga , Femenino , Flumazenil/farmacología , Agonistas de Receptores de GABA-A , Ratas , Ratas Wistar
15.
Epilepsy Res ; 18(2): 91-6, 1994 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-7957040

RESUMEN

An earlier pharmacodynamic study of the chiral antiepileptic drug stiripentol in an intravenous pentylenetetrazol-induced seizure model in the rat showed the development of a significant degree of tolerance to the anticonvulsant and neurotoxic effects following subacute treatment with the racemic compound. A more recent study with the pure enantiomers of stiripentol indicated that the (+)-enantiomer is 2.4 times more potent than the (-)-enantiomer, based on a comparison of brain EC50 values for the anticonvulsant effect. Moreover, (-)-stiripentol has a much longer elimination half-life than (+)-stiripentol. We have re-analyzed the brain and blood samples from the first pharmacodynamic study using a newly developed chiral HPLC assay to investigate whether the tolerance phenomenon with racemic stiripentol was due to a shift in the enantiomeric composition of stiripentol in brain tissue during repetitive administration of racemic drug. A large increase, as much as 5-6-fold, in the (-)/(+) ratio in brain concentration of stiripentol was observed after subacute administration, as compared with that after a single dose of the racemic drug. The enrichment in the less potent enantiomers during repetitive drug administration explains the previous observation of an apparent development of tolerance when the pharmacologic effects were related to total [(-)+(+)] brain concentrations of stiripentol as measured by a non-stereoselective assay. The results of this study highlight the importance of stereoselective pharmacokinetics in investigating the pharmacodynamics of the racemic mixture of a chiral anticonvulsant.


Asunto(s)
Anticonvulsivantes/farmacocinética , Dioxolanos/farmacocinética , Animales , Anticonvulsivantes/química , Anticonvulsivantes/farmacología , Encéfalo/metabolismo , Cromatografía Líquida de Alta Presión , Dioxolanos/química , Dioxolanos/farmacología , Tolerancia a Medicamentos , Semivida , Técnicas In Vitro , Pentilenotetrazol , Ratas , Convulsiones/inducido químicamente , Convulsiones/prevención & control , Estereoisomerismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...