Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
N Engl J Med ; 389(4): 309-321, 2023 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-37494485

RESUMEN

BACKGROUND: Narcolepsy type 1 is caused by severe loss or lack of brain orexin neuropeptides. METHODS: We conducted a phase 2, randomized, placebo-controlled trial of TAK-994, an oral orexin receptor 2-selective agonist, in patients with narcolepsy type 1. Patients with confirmed narcolepsy type 1 according to clinical criteria were randomly assigned to receive twice-daily oral TAK-994 (30 mg, 90 mg, or 180 mg) or placebo. The primary end point was the mean change from baseline to week 8 in average sleep latency (the time it takes to fall asleep) on the Maintenance of Wakefulness Test (range, 0 to 40 minutes; normal ability to stay awake, ≥20 minutes). Secondary end points included the change in the Epworth Sleepiness Scale (ESS) score (range, 0 to 24, with higher scores indicating greater daytime sleepiness; normal, <10) and the weekly cataplexy rate. RESULTS: Of the 73 patients, 17 received TAK-994 at a dose of 30 mg twice daily, 20 received 90 mg twice daily, 19 received 180 mg twice daily, and 17 received placebo. The phase 2 trial and an extension trial were terminated early owing to hepatic adverse events. Primary end-point data were available for 41 patients (56%); the main reason for missing data was early trial termination. Least-squares mean changes to week 8 in average sleep latency on the MWT were 23.9 minutes in the 30-mg group, 27.4 minutes in the 90-mg group, 32.6 minutes in the 180-mg group, and -2.5 minutes in the placebo group (difference vs. placebo, 26.4 minutes in the 30-mg group, 29.9 minutes in the 90-mg group, and 35.0 minutes the 180-mg group; P<0.001 for all comparisons). Least-squares mean changes to week 8 in the ESS score were -12.2 in the 30-mg group, -13.5 in the 90-mg group, -15.1 in the 180-mg group, and -2.1 in the placebo group (difference vs. placebo, -10.1 in the 30-mg group, -11.4 in the 90-mg group, and -13.0 in the 180-mg group). Weekly incidences of cataplexy at week 8 were 0.27 in the 30-mg group, 1.14 in the 90-mg group, 0.88 in the 180-mg group, and 5.83 in the placebo group (rate ratio vs. placebo, 0.05 in the 30-mg group, 0.20 in the 90-mg group, and 0.15 in the 180-mg group). A total of 44 of 56 patients (79%) receiving TAK-994 had adverse events, most commonly urinary urgency or frequency. Clinically important elevations in liver-enzyme levels occurred in 5 patients, and drug-induced liver injury meeting Hy's law criteria occurred in 3 patients. CONCLUSIONS: In a phase 2 trial involving patients with narcolepsy type 1, an orexin receptor 2 agonist resulted in greater improvements on measures of sleepiness and cataplexy than placebo over a period of 8 weeks but was associated with hepatotoxic effects. (Funded by Takeda Development Center Americas; TAK-994-1501 and TAK-994-1504 ClinicalTrials.gov numbers, NCT04096560 and NCT04820842.).


Asunto(s)
Narcolepsia , Receptores de Orexina , Orexinas , Humanos , Cataplejía/complicaciones , Cataplejía/tratamiento farmacológico , Cataplejía/epidemiología , Método Doble Ciego , Narcolepsia/tratamiento farmacológico , Narcolepsia/complicaciones , Narcolepsia/epidemiología , Receptores de Orexina/agonistas , Receptores de Orexina/uso terapéutico , Somnolencia/efectos de los fármacos , Resultado del Tratamiento , Orexinas/análisis , Orexinas/deficiencia , Orexinas/farmacología , Química Encefálica/efectos de los fármacos , Administración Oral , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología
2.
Ann Neurol ; 93(3): 604-614, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36401339

RESUMEN

OBJECTIVE: The radiologically isolated syndrome (RIS) represents the earliest detectable pre-clinical phase of multiple sclerosis (MS). This study evaluated the impact of therapeutic intervention in preventing first symptom manifestation at this stage in the disease spectrum. METHODS: We conducted a multi-center, randomized, double-blinded, placebo-controlled study involving people with RIS. Individuals without clinical symptoms typical of MS but with incidental brain MRI anomalies consistent with central nervous system (CNS) demyelination were included. Within 12 MS centers in the United States, participants were randomly assigned 1:1 to oral dimethyl fumarate (DMF) 240 mg twice daily or placebo. The primary endpoint was the time to onset of clinical symptoms attributable to a CNS demyelinating event within a follow-up period of 96 weeks. An intention-to-treat analysis was applied to all participating individuals in the primary and safety investigations. The study is registered at ClinicalTrials.gov, NCT02739542 (ARISE). RESULTS: Participants from 12 centers were recruited from March 9, 2016, to October 31, 2019, with 44 people randomized to dimethyl fumarate and 43 to placebo. Following DMF treatment, the risk of a first clinical demyelinating event during the 96-week study period was highly reduced in the unadjusted Cox proportional-hazards regression model (hazard ratio [HR] = 0.18, 95% confidence interval [CI] = 0.05-0.63, p = 0.007). More moderate adverse reactions were present in the DMF (34 [32%]) than placebo groups (19 [21%]) but severe events were similar (DMF, 3 [5%]; placebo, 4 [9%]). INTERPRETATION: This is the first randomized clinical trial demonstrating the benefit of a disease-modifying therapy in preventing a first acute clinical event in people with RIS. ANN NEUROL 2023;93:604-614.


Asunto(s)
Esclerosis Múltiple Recurrente-Remitente , Esclerosis Múltiple , Humanos , Dimetilfumarato/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Inmunosupresores/uso terapéutico , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Imagen por Resonancia Magnética , Método Doble Ciego
3.
Alzheimers Dement ; 20(5): 3406-3415, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38567735

RESUMEN

INTRODUCTION: Aducanumab selectively targets aggregated forms of amyloid beta (Aß), a neuropathological hallmark of Alzheimer's disease (AD). METHODS: PRIME was a Phase 1b, double-blind, randomized clinical trial of aducanumab. During the 12-month placebo-controlled period, participants with prodromal AD or mild AD dementia were randomized to receive aducanumab or placebo. At week 56, participants could enroll in a long-term extension (LTE), in which all participants received aducanumab. The primary endpoint was safety and tolerability. RESULTS: Amyloid-related imaging abnormalities-edema (ARIA-E) were the most common adverse event. Dose titration was associated with a decrease in the incidence of ARIA-E. Over 48 months, aducanumab decreased brain amyloid levels in a dose- and time-dependent manner. Exploratory endpoints suggested a continued benefit in the reduction of clinical decline over 48 months. DISCUSSION: The safety profile of aducanumab remained unchanged in the LTE of PRIME. Amyloid plaque levels continued to decrease in participants treated with aducanumab. HIGHLIGHTS: PRIME was a Phase 1b, double-blind, randomized clinical trial of aducanumab. We report cumulative safety and 48-month efficacy results from PRIME. Amyloid-related imaging abnormalities-edema (ARIA-E) were the most common adverse event (AE); 61% of participants with ARIA-E were asymptomatic. Dose titration was associated with a decrease in the incidence of ARIA-E. Aducanumab decreased levels of amyloid beta (Aß) in a dose- and time-dependent manner.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Anticuerpos Monoclonales Humanizados , Humanos , Método Doble Ciego , Anticuerpos Monoclonales Humanizados/uso terapéutico , Enfermedad de Alzheimer/tratamiento farmacológico , Masculino , Femenino , Anciano , Péptidos beta-Amiloides/metabolismo , Encéfalo/diagnóstico por imagen , Encéfalo/efectos de los fármacos , Encéfalo/patología , Resultado del Tratamiento , Placa Amiloide/tratamiento farmacológico , Relación Dosis-Respuesta a Droga
4.
Acta Neuropathol ; 144(1): 143-153, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35581440

RESUMEN

Amyloid beta (Aß) plaque is a defining pathologic feature of Alzheimer disease (AD). Aducanumab, a monoclonal IgG1 that selectively binds aggregated species of Aß, has been shown by amyloid positron emission tomography (Amyloid PET) to reduce Aß plaques in patients with prodromal and mild AD. This is the first autopsy report of the AD neuropathology in a patient previously treated with aducanumab. The patient was an 84-year-old woman who was randomized to the placebo arm of the PRIME Phase 1b study (221AD103). The patient progressed to moderate dementia (MMSE = 14/30), beyond the targeted early AD treatment stage, before receiving aducanumab in the long-term extension (LTE). The patient then received 32 monthly doses of aducanumab, titrated up to 6 mg/kg, for a cumulative dose of 186 mg/kg. In the LTE, Amyloid PET scans demonstrated robust Aß plaque reduction, from a composite standard uptake value ratio (SUVR) of 1.5 at screening to < 1.1 at 56 weeks post-aducanumab dosing. MRI examinations were negative for amyloid-related imaging abnormalities (ARIA). She passed away in hospice care 4 months after her last dose of aducanumab. The postmortem neuropathologic examination confirmed AD neuropathologic changes. Aß and IBA1 immunohistochemistry assays demonstrated sparse residual Aß plaque engaged by amoeboid reactive microglia. Phospho-Tau (pTau) immunohistochemistry demonstrated neocortical neurofibrillary degeneration (Braak stage V, NIA/AA Stage B3). However, the density of pTau neuropathology, including neuritic plaque pTau (NP-Tau), appeared lower in the PRIME LTE Patient compared to a reference cohort of untreated Braak stage V-VI, NIA/AA Stage B3 AD cases. Taken together, this case report is the first to provide Amyloid PET and neuropathologic evidence substantiating the impact of aducanumab to reduce Aß plaque neuropathology in a patient with AD. Furthermore, this report underscores the critical importance of autopsy neuropathology studies to augment our understanding of aducanumab's mechanism of action and impact on AD biomarkers.


Asunto(s)
Enfermedad de Alzheimer , Anticuerpos Monoclonales Humanizados , Anciano de 80 o más Años , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Anticuerpos Monoclonales Humanizados/uso terapéutico , Ensayos Clínicos Fase I como Asunto , Femenino , Humanos , Ovillos Neurofibrilares/patología , Placa Amiloide/patología , Placa Amiloide/prevención & control , Tomografía de Emisión de Positrones , Ensayos Clínicos Controlados Aleatorios como Asunto
5.
Nature ; 501(7465): 52-7, 2013 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-23965627

RESUMEN

Nociceptor sensory neurons are specialized to detect potentially damaging stimuli, protecting the organism by initiating the sensation of pain and eliciting defensive behaviours. Bacterial infections produce pain by unknown molecular mechanisms, although they are presumed to be secondary to immune activation. Here we demonstrate that bacteria directly activate nociceptors, and that the immune response mediated through TLR2, MyD88, T cells, B cells, and neutrophils and monocytes is not necessary for Staphylococcus aureus-induced pain in mice. Mechanical and thermal hyperalgesia in mice is correlated with live bacterial load rather than tissue swelling or immune activation. Bacteria induce calcium flux and action potentials in nociceptor neurons, in part via bacterial N-formylated peptides and the pore-forming toxin α-haemolysin, through distinct mechanisms. Specific ablation of Nav1.8-lineage neurons, which include nociceptors, abrogated pain during bacterial infection, but concurrently increased local immune infiltration and lymphadenopathy of the draining lymph node. Thus, bacterial pathogens produce pain by directly activating sensory neurons that modulate inflammation, an unsuspected role for the nervous system in host-pathogen interactions.


Asunto(s)
Inflamación/microbiología , Nociceptores/metabolismo , Dolor/microbiología , Dolor/fisiopatología , Staphylococcus aureus/patogenicidad , Potenciales de Acción , Animales , Carga Bacteriana , Señalización del Calcio , Femenino , Proteínas Hemolisinas/metabolismo , Interacciones Huésped-Patógeno , Calor , Hiperalgesia/microbiología , Inmunidad Innata , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Enfermedades Linfáticas/inmunología , Enfermedades Linfáticas/microbiología , Enfermedades Linfáticas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos , Factor 88 de Diferenciación Mieloide/inmunología , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Canal de Sodio Activado por Voltaje NAV1.8/deficiencia , Canal de Sodio Activado por Voltaje NAV1.8/inmunología , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Neutrófilos , Dolor/inmunología , Dolor/metabolismo , Estabilidad Proteica , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/inmunología , Staphylococcus aureus/metabolismo , Receptor Toll-Like 2/inmunología
6.
J Neurosci ; 32(18): 6364-72, 2012 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-22553041

RESUMEN

Epoxyeicosatrienoic acids (EETs) are cytochrome P450-epoxygenase-derived metabolites of arachidonic acid that act as endogenous signaling molecules in multiple biological systems. Here we have investigated the specific contribution of 5,6-EET to transient receptor potential (TRP) channel activation in nociceptor neurons and its consequence for nociceptive processing. We found that, during capsaicin-induced nociception, 5,6-EET levels increased in dorsal root ganglia (DRGs) and the dorsal spinal cord, and 5,6-EET is released from activated sensory neurons in vitro. 5,6-EET potently induced a calcium flux (100 nm) in cultured DRG neurons that was completely abolished when TRPA1 was deleted or inhibited. In spinal cord slices, 5,6-EET dose dependently enhanced the frequency, but not the amplitude, of spontaneous EPSCs (sEPSCs) in lamina II neurons that also responded to mustard oil (allyl isothiocyanate), indicating a presynaptic action. Furthermore, 5,6-EET-induced enhancement of sEPSC frequency was abolished in TRPA1-null mice, suggesting that 5,6-EET presynaptically facilitated spinal cord synaptic transmission by TRPA1. Finally, in vivo intrathecal injection of 5,6-EET caused mechanical allodynia in wild-type but not TRPA1-null mice. We conclude that 5,6-EET is synthesized on the acute activation of nociceptors and can produce mechanical hypersensitivity via TRPA1 at central afferent terminals in the spinal cord.


Asunto(s)
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Potenciales de Acción/fisiología , Vías Aferentes/fisiopatología , Hiperalgesia/fisiopatología , Células Receptoras Sensoriales/metabolismo , Ácido 8,11,14-Eicosatrienoico/metabolismo , Animales , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
7.
J Clin Invest ; 118(5): 1899-910, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18398506

RESUMEN

Sensory neurons in the airways are finely tuned to respond to reactive chemicals threatening airway function and integrity. Nasal trigeminal nerve endings are particularly sensitive to oxidants formed in polluted air and during oxidative stress as well as to chlorine, which is frequently released in industrial and domestic accidents. Oxidant activation of airway neurons induces respiratory depression, nasal obstruction, sneezing, cough, and pain. While normally protective, chemosensory airway reflexes can provoke severe complications in patients affected by inflammatory airway conditions like rhinitis and asthma. Here, we showed that both hypochlorite, the oxidizing mediator of chlorine, and hydrogen peroxide, a reactive oxygen species, activated Ca(2+) influx and membrane currents in an oxidant-sensitive subpopulation of chemosensory neurons. These responses were absent in neurons from mice lacking TRPA1, an ion channel of the transient receptor potential (TRP) gene family. TRPA1 channels were strongly activated by hypochlorite and hydrogen peroxide in primary sensory neurons and heterologous cells. In tests of respiratory function, Trpa1(-/-) mice displayed profound deficiencies in hypochlorite- and hydrogen peroxide-induced respiratory depression as well as decreased oxidant-induced pain behavior. Our results indicate that TRPA1 is an oxidant sensor in sensory neurons, initiating neuronal excitation and subsequent physiological responses in vitro and in vivo.


Asunto(s)
Neuronas Aferentes/metabolismo , Oxidantes/metabolismo , Sistema Respiratorio , Canales de Potencial de Receptor Transitorio/metabolismo , Animales , Calcio/metabolismo , Línea Celular , Humanos , Peróxido de Hidrógeno/metabolismo , Potenciales de la Membrana/fisiología , Ratones , Ratones Noqueados , Planta de la Mostaza/metabolismo , Neuronas Aferentes/citología , Dolor/inducido químicamente , Dolor/metabolismo , Técnicas de Placa-Clamp , Aceites de Plantas/metabolismo , Sistema Respiratorio/citología , Sistema Respiratorio/metabolismo , Hipoclorito de Sodio/metabolismo , Canal Catiónico TRPA1 , Canales de Potencial de Receptor Transitorio/genética
8.
FASEB J ; 23(4): 1102-14, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19036859

RESUMEN

The release of methyl isocyanate in Bhopal, India, caused the worst industrial accident in history. Exposures to industrial isocyanates induce lacrimation, pain, airway irritation, and edema. Similar responses are elicited by chemicals used as tear gases. Despite frequent exposures, the biological targets of isocyanates and tear gases in vivo have not been identified, precluding the development of effective countermeasures. We use Ca(2+) imaging and electrophysiology to show that the noxious effects of isocyanates and those of all major tear gas agents are caused by activation of Ca(2+) influx and membrane currents in mustard oil-sensitive sensory neurons. These responses are mediated by transient receptor potential ankyrin 1 (TRPA1), an ion channel serving as a detector for reactive chemicals. In mice, genetic ablation or pharmacological inhibition of TRPA1 dramatically reduces isocyanate- and tear gas-induced nocifensive behavior after both ocular and cutaneous exposures. We conclude that isocyanates and tear gas agents target the same neuronal receptor, TRPA1. Treatment with TRPA1 antagonists may prevent and alleviate chemical irritation of the eyes, skin, and airways and reduce the adverse health effects of exposures to a wide range of toxic noxious chemicals.


Asunto(s)
Isocianatos/toxicidad , Gases Lacrimógenos/toxicidad , Canales de Potencial de Receptor Transitorio/antagonistas & inhibidores , Animales , Células CHO , Línea Celular , Células Cultivadas , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Electrofisiología , Femenino , Ganglios Espinales/citología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Ácido Hipocloroso/farmacología , Riñón/citología , Riñón/embriología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas Aferentes/citología , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/metabolismo , Oxidantes/farmacología , Técnicas de Placa-Clamp , Canal Catiónico TRPA1 , Canales de Potencial de Receptor Transitorio/genética , Ganglio del Trigémino/citología , Ganglio del Trigémino/efectos de los fármacos , Ganglio del Trigémino/metabolismo
9.
Neurology ; 92(15): e1724-e1738, 2019 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-30918100

RESUMEN

OBJECTIVE: To assess functional changes in lymphocyte repertoire and subsequent clinical implications during delayed-release dimethyl fumarate (DMF) treatment in patients with multiple sclerosis. METHODS: Using peripheral blood from several clinical trials of DMF, immune cell subsets were quantified using flow cytometry. For some patients, lymphocyte counts were assessed after DMF discontinuation. Incidence of adverse events, including serious and opportunistic infections, was assessed. RESULTS: In DMF-treated patients, absolute lymphocyte counts (ALCs) demonstrated a pattern of decline followed by stabilization, which also was reflected in the global reduction in numbers of circulating functional lymphocyte subsets. The relative frequencies of circulating memory T- and B-cell populations declined and naive cells increased. No increased incidence of serious infection or malignancy was observed for patients treated with DMF, even when stratified by ALC or T-cell subset frequencies. For patients who discontinued DMF due to lymphopenia, ALCs increased after DMF discontinuation; recovery time varied by ALC level at discontinuation. T-cell subsets closely correlated with ALCs in both longitudinal and cross-sectional analyses. CONCLUSIONS: DMF shifted the immunophenotype of circulating lymphocyte subsets. ALCs were closely correlated with CD4+ and CD8+ T-cell counts, indicating that lymphocyte subset monitoring is not required for safety vigilance. No increased risk of serious infection was observed in patients with low T-cell subset counts. Monitoring ALC remains the most effective way of identifying patients at risk of subsequently developing prolonged moderate to severe lymphopenia, a risk factor for progressive multifocal leukoencephalopathy in DMF-treated patients. TRIAL REGISTRATION NUMBERS: EUDRA CT 2015-001973-42, NCT00168701, NCT00420212, NCT00451451, and NCT00835770.


Asunto(s)
Dimetilfumarato/uso terapéutico , Inmunosupresores/uso terapéutico , Linfocitos/efectos de los fármacos , Esclerosis Múltiple Recurrente-Remitente/sangre , Adulto , Linfocitos B/efectos de los fármacos , Relación CD4-CD8 , Estudios Transversales , Preparaciones de Acción Retardada , Dimetilfumarato/efectos adversos , Dimetilfumarato/farmacología , Femenino , Humanos , Inmunosupresores/efectos adversos , Inmunosupresores/farmacología , Estudios Longitudinales , Recuento de Linfocitos , Linfopenia/sangre , Linfopenia/inducido químicamente , Masculino , Persona de Mediana Edad , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Esclerosis Múltiple Recurrente-Remitente/inmunología , Medición de Riesgo , Linfocitos T/efectos de los fármacos
10.
J Physiol ; 586(21): 5161-79, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18787033

RESUMEN

The rates of activation and unitary properties of Na+-activated K+ (K(Na)) currents have been found to vary substantially in different types of neurones. One class of K(Na) channels is encoded by the Slack gene. We have now determined that alternative RNA splicing gives rise to at least five different transcripts for Slack, which produce Slack channels that differ in their predicted cytoplasmic amino-termini and in their kinetic properties. Two of these, termed Slack-A channels, contain an amino-terminus domain closely resembling that of another class of K(Na) channels encoded by the Slick gene. Neuronal expression of Slack-A channels and of the previously described Slack isoform, now called Slack-B, are driven by independent promoters. Slack-A mRNAs were enriched in the brainstem and olfactory bulb and detected at significant levels in four different brain regions. When expressed in CHO cells, Slack-A channels activate rapidly upon depolarization and, in single channel recordings in Xenopus oocytes, are characterized by multiple subconductance states with only brief transient openings to the fully open state. In contrast, Slack-B channels activate slowly over hundreds of milliseconds, with openings to the fully open state that are approximately 6-fold longer than those for Slack-A channels. In numerical simulations, neurones in which outward currents are dominated by a Slack-A-like conductance adapt very rapidly to repeated or maintained stimulation over a wide range of stimulus strengths. In contrast, Slack-B currents promote rhythmic firing during maintained stimulation, and allow adaptation rate to vary with stimulus strength. Using an antibody that recognizes all amino-termini isoforms of Slack, Slack immunoreactivity is present at locations that have no Slack-B-specific staining, including olfactory bulb glomeruli and the dendrites of hippocampal neurones, suggesting that Slack channels with alternate amino-termini such as Slack-A channels are present at these locations. Our data suggest that alternative promoters of the Slack gene differentially modulate the properties of neurones.


Asunto(s)
Potenciales de Acción/fisiología , Adaptación Fisiológica/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/fisiología , Canales de Potasio/metabolismo , Secuencia de Aminoácidos , Animales , Encéfalo/metabolismo , Clonación Molecular , Regulación de la Expresión Génica/fisiología , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Canales de Potasio/genética , Canales de potasio activados por Sodio , Regiones Promotoras Genéticas , Isoformas de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas
11.
Neurol Neuroimmunol Neuroinflamm ; 5(1): e409, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29159204

RESUMEN

OBJECTIVES: To investigate the immune response to vaccinations in patients with relapsing forms of MS treated with delayed-release dimethyl fumarate (DMF) vs nonpegylated interferon (IFN). METHODS: In this open-label, multicenter study, patients received 3 vaccinations: (1) tetanus-diphtheria toxoid (Td) to test T-cell-dependent recall response, (2) pneumococcal vaccine polyvalent to test T-cell-independent humoral response, and (3) meningococcal (groups A, C, W-135, and Y) oligosaccharide CRM197 conjugate to test T-cell-dependent neoantigen response. Eligible patients were aged 18-55 years, diagnosed with relapsing-remitting MS (RRMS), and either treated for ≥6 months with an approved dose of DMF or for ≥3 months with an approved dose of nonpegylated IFN. Primary end point was the proportion of patients with ≥2-fold rise in antitetanus serum IgG levels from prevaccination to 4 weeks after vaccination. RESULTS: Seventy-one patients (DMF treated, 38; IFN treated, 33) were enrolled. The mean age was 45.3 years (range 27-55); 86% were women. Responder rates (≥2-fold rise) to Td vaccination were comparable between DMF- and IFN-treated groups (68% vs 73%). Responder rates (≥2-fold rise) were also similar between DMF- and IFN-treated groups for diphtheria antitoxoid (58% vs 61%), pneumococcal serotype 3 (66% vs 79%), pneumococcal serotype 8 (95% vs 88%), and meningococcal serogroup C (53% vs 53%), all p > 0.05. In a post hoc analysis, no meaningful differences were observed between groups in the proportion of responders when stratified by age category or lymphocyte count. CONCLUSIONS: DMF-treated patients mount an immune response to recall, neoantigens, and T-cell-independent antigens, which was comparable with that of IFN-treated patients and provided adequate seroprotection. CLINICALTRIALSGOV IDENTIFIER: NCT02097849. CLASSIFICATION OF EVIDENCE: This study provides Class II evidence that patients with RRMS treated with DMF respond to vaccinations comparably with IFN-treated patients.

12.
Int J MS Care ; 18(1): 9-18, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26917993

RESUMEN

BACKGROUND: In phase 3 trials, delayed-release dimethyl fumarate (DMF; also known as gastroresistant DMF) demonstrated efficacy in relapsing-remitting multiple sclerosis (MS). Gastrointestinal (GI) events were associated with DMF treatment. The single-arm, open-label MANAGE study examined the incidence, severity, duration, and management of GI events in adults with relapsing MS initiating DMF treatment in clinical practice in the United States shortly after marketing approval. PATIENTS AND METHODS: Patients (N = 233) took DMF for up to 12 weeks and recorded information regarding GI events using an eDiary and numerical rating scales. RESULTS: Overall, 54.1% of patients used symptomatic therapy and had GI symptoms. The incidence of GI events was highest in the first month of treatment. The duration of GI events varied by event type, and severity was generally mild to moderate. Decreased severity was seen in patients treated with antacids, bismuth subsalicylate, acid-secretion blockers, antidiarrheals, and antiemetics. Less than 10% of patients were using symptomatic therapy for GI events by week 12 of DMF treatment. A modest reduction in severe GI events was observed in patients who regularly took DMF with food compared with patients who did not. The incidence of GI-related events was comparable in patients with or without a history of GI abnormalities and in patients who did or did not use alcohol or tobacco. CONCLUSIONS: Gastrointestinal events associated with DMF are generally transient, mild to moderate in severity, and manageable. Symptomatic therapy and dosing with food may mitigate these events.

13.
J Neurosci ; 24(8): 1936-40, 2004 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-14985434

RESUMEN

The promoter for the kv3.1 potassium channel gene is regulated by a Ca2+-cAMP responsive element, which binds the transcription factor cAMP response element-binding protein (CREB). Kv3.1 is expressed in a tonotopic gradient within the medial nucleus of the trapezoid body (MNTB) of the auditory brainstem, where Kv3.1 levels are highest at the medial end, which corresponds to high auditory frequencies. We have compared the levels of Kv3.1, CREB, and the phosphorylated form of CREB (pCREB) in a mouse strain that maintains good hearing throughout life, CBA/J (CBA), with one that suffers early cochlear hair cell loss, C57BL/6 (BL/6). A gradient of Kv3.1 immunoreactivity in the MNTB was detected in both young (6 week) and older (8 month) CBA mice. Although no gradient of CREB was detected, pCREB-immunopositive cells were grouped together in distinct clusters along the tonotopic axis. The same pattern of Kv3.1, CREB, and pCREB localization was also found in young BL/6 mice at a time (6 weeks) when hearing is normal. In contrast, at 8 months, when hearing is impaired, the gradient of Kv3.1 was abolished. Moreover, in the older BL/6 mice there was a decrease in CREB expression along the tonotopic axis, and the pattern of pCREB labeling appeared random, with no discrete clusters of pCREB-positive cells along the tonotopic axis. Our findings are consistent with the hypothesis that ongoing activity in auditory brainstem neurons is necessary for the maintenance of Kv3.1 tonotopicity through the CREB pathway.


Asunto(s)
Vías Auditivas/fisiopatología , Tronco Encefálico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Neuronas/metabolismo , Neuropéptidos/metabolismo , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/metabolismo , Presbiacusia/fisiopatología , Estimulación Acústica , Factores de Edad , Animales , Vías Auditivas/patología , Tronco Encefálico/patología , Cerebelo/metabolismo , Cerebelo/patología , Progresión de la Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Endogámicos DBA , Neuropéptidos/genética , Fosforilación , Canales de Potasio/genética , Presbiacusia/patología , Reflejo de Sobresalto/fisiología , Canales de Potasio Shaw
14.
J Neurosci ; 23(4): 1133-41, 2003 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-12598601

RESUMEN

Neurons of the medial nucleus of the trapezoid body, which transmit auditory information that is used to compute the location of sounds in space, are capable of firing at high frequencies with great temporal precision. We found that elimination of the Kv3.1 gene in mice results in the loss of a high-threshold component of potassium current and failure of the neurons to follow high-frequency stimulation. A partial decrease in Kv3.1 current can be produced in wild-type neurons of the medial nucleus of the trapezoid body by activation of protein kinase C. Paradoxically, activation of protein kinase C increases temporal fidelity and the number of action potentials that are evoked by intermediate frequencies of stimulation. Computer simulations confirm that a partial decrease in Kv3.1 current is sufficient to increase the accuracy of response at intermediate frequencies while impairing responses at high frequencies. We further establish that, of the two isoforms of the Kv3.1 potassium channel that are expressed in these neurons, Kv3.1a and Kv3.1b, the decrease in Kv3.1 current is mediated by selective phosphorylation of the Kv3.1b isoform. Using site-directed mutagenesis, we identify a specific C-terminal phosphorylation site responsible for the observed difference in response of the two isoforms to protein kinase C activation. Our results suggest that modulation of Kv3.1 by phosphorylation allows auditory neurons to tune their responses to different patterns of sensory stimulation.


Asunto(s)
Potenciales de Acción , Tronco Encefálico/fisiología , Potenciales Evocados Auditivos , Neuronas/fisiología , Neuropéptidos/metabolismo , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/metabolismo , Animales , Tronco Encefálico/citología , Células CHO , Células Cultivadas , Cricetinae , Conductividad Eléctrica , Cinética , Ratones , Ratones Noqueados , Neuronas/metabolismo , Neuropéptidos/genética , Neuropéptidos/fisiología , Técnicas de Placa-Clamp , Fosforilación , Canales de Potasio/genética , Canales de Potasio/fisiología , Isoformas de Proteínas/metabolismo , Proteína Quinasa C/metabolismo , Serina/metabolismo , Canales de Potasio Shaw , Acetato de Tetradecanoilforbol/farmacología
15.
J Comp Neurol ; 484(1): 80-92, 2005 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-15717307

RESUMEN

Na+-activated K+ currents (K(Na)) have been reported in multiple neuronal nuclei and the properties of K(Na) vary in different cell types. We have described previously the distribution of Slack, a Na+-activated K+ channel subunit. Another recently cloned Na+-activated K+ channel is Slick, which differs from Slack in its rapid activation and its sensitivity to intracellular ATP levels. We now report the localization of Slick in the rat central nervous system using in situ and immunohistochemical techniques. As for Slack, we find that Slick is widely distributed in the brain. Specifically, strong hybridization signals and immunoreactivity were found in the brainstem, including auditory neurons such as the medial nucleus of the trapezoid body. As has also been shown for Slack, Slick is expressed in the olfactory bulb, red nucleus, facial nucleus, pontine nucleus, oculomotor nucleus, substantia nigra, deep cerebellar nuclei, vestibular nucleus, and the thalamus. Slick mRNA and protein, however, also are found in certain neurons that do not express Slack. These neurons include those of the hippocampal CA1, CA2, and CA3 regions, the dentate gyrus, supraoptic nucleus, hypothalamus, and cortical layers II, III, and V. These data suggest that Slick may function independently of Slack in these regions. Computer simulations indicate that Slick currents can cause adaptation during prolonged stimuli. Such adaptation allows a neuron to respond to high-frequency stimulation with lower-frequency firing that remains temporally locked to individual stimuli, a property seen in many auditory neurons. Although it is not yet known if Slick and Slack subunits heteromultimerize, the existence of two genes that encode K(Na), that are widely expressed in the nervous system, with both overlapping and nonoverlapping distributions, provides the basis for the reported heterogeneity in the properties of K(Na) from various neurons.


Asunto(s)
Sistema Nervioso Central/anatomía & histología , Sistema Nervioso Central/metabolismo , Canales de Potasio/metabolismo , Animales , Vías Auditivas/anatomía & histología , Vías Auditivas/metabolismo , Vías Auditivas/ultraestructura , Células CHO , Simulación por Computador , Cricetinae , ADN Complementario/genética , Nervio Facial/anatomía & histología , Nervio Facial/metabolismo , Nervio Facial/ultraestructura , Immunoblotting , Inmunohistoquímica , Hibridación in Situ , Cinética , Modelos Neurológicos , Neuronas/metabolismo , Neuronas/ultraestructura , Bulbo Olfatorio/metabolismo , Canales de potasio activados por Sodio , Sondas ARN , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Fracciones Subcelulares/metabolismo , Fracciones Subcelulares/ultraestructura
16.
Hear Res ; 206(1-2): 133-45, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16081004

RESUMEN

The firing patterns of neurons in central auditory pathways encode specific features of sound stimuli, such as frequency, intensity and localization in space. The generation of the appropriate pattern depends, to a major extent, on the properties of the voltage-dependent potassium channels in these neurons. The mammalian auditory pathways that compute the direction of a sound source are located in the brainstem and include the connection from bushy cells in the anteroventral cochlear nucleus (AVCN) to the principal neurons of the medial nucleus of the trapezoid body (MNTB). To preserve the fidelity of timing of action potentials that is required for sound localization, these neurons express several types of potassium channels, including the Kv3 and Kv1 families of voltage-dependent channels and the Slick and Slack sodium-dependent channels. These channels determine the pattern of action potentials and the amount of neurotransmitter released during repeated stimulation. The amplitude of currents carried by one of these channels, the Kv3.1b channel, is regulated in the short term by protein phosphorylation, and in the long term, by changes in gene expression, such that the intrinsic excitability of the neurons is constantly being regulated by the ambient auditory environment.


Asunto(s)
Potenciales de Acción/fisiología , Vías Auditivas/fisiología , Nervio Coclear/fisiología , Neuronas/fisiología , Canales de Potasio/metabolismo , Núcleos Vestibulares/fisiología , Animales , Núcleo Coclear/fisiología , Humanos , Fosforilación , Canales de Potasio/genética , Proteínas Quinasas/metabolismo , ARN Mensajero/metabolismo , Tiempo de Reacción , Sinapsis/metabolismo , Factores de Tiempo
17.
Neuron ; 87(2): 341-54, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26119026

RESUMEN

Lung nociceptors initiate cough and bronchoconstriction. To elucidate if these fibers also contribute to allergic airway inflammation, we stimulated lung nociceptors with capsaicin and observed increased neuropeptide release and immune cell infiltration. In contrast, ablating Nav1.8(+) sensory neurons or silencing them with QX-314, a charged sodium channel inhibitor that enters via large-pore ion channels to specifically block nociceptors, substantially reduced ovalbumin- or house-dust-mite-induced airway inflammation and bronchial hyperresponsiveness. We also discovered that IL-5, a cytokine produced by activated immune cells, acts directly on nociceptors to induce the release of vasoactive intestinal peptide (VIP). VIP then stimulates CD4(+) and resident innate lymphoid type 2 cells, creating an inflammatory signaling loop that promotes allergic inflammation. Our results indicate that nociceptors amplify pathological adaptive immune responses and that silencing these neurons with QX-314 interrupts this neuro-immune interplay, revealing a potential new therapeutic strategy for asthma.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/inmunología , Nociceptores/fisiología , Hipersensibilidad Respiratoria/inmunología , Anestésicos Locales/farmacología , Animales , Animales Recién Nacidos , Capsaicina/farmacología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Adyuvante de Freund/toxicidad , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Interleucina-5/metabolismo , Lidocaína/análogos & derivados , Lidocaína/farmacología , Ratones , Nociceptores/efectos de los fármacos , Nociceptores/metabolismo , Ovalbúmina/toxicidad , Hipersensibilidad Respiratoria/inducido químicamente , Factores de Tiempo , Péptido Intestinal Vasoactivo/metabolismo
18.
Neuron ; 73(4): 638-52, 2012 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-22365541

RESUMEN

After nerve injury maladaptive changes can occur in injured sensory neurons and along the entire nociceptive pathway within the CNS, which may lead to spontaneous pain or pain hypersensitivity. The resulting neuropathic pain syndromes present as a complex combination of negative and positive symptoms, which vary enormously from individual to individual. This variation depends on a diversity of underlying pathophysiological changes resulting from the convergence of etiological, genotypic, and environmental factors. The pain phenotype can serve therefore, as a window on underlying pathophysiological neural mechanisms and as a guide for developing personalized pain medicine.


Asunto(s)
Vías Aferentes , Neuralgia/diagnóstico , Neuralgia/fisiopatología , Nervios Periféricos/patología , Fenotipo , Vías Aferentes/patología , Vías Aferentes/fisiopatología , Humanos , Modelos Biológicos , Neuralgia/etiología , Neuralgia/genética , Dimensión del Dolor , Umbral del Dolor/fisiología
19.
Nat Neurosci ; 15(8): 1063-7, 2012 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-22837035

RESUMEN

The peripheral nervous and immune systems are traditionally thought of as serving separate functions. The line between them is, however, becoming increasingly blurred by new insights into neurogenic inflammation. Nociceptor neurons possess many of the same molecular recognition pathways for danger as immune cells, and, in response to danger, the peripheral nervous system directly communicates with the immune system, forming an integrated protective mechanism. The dense innervation network of sensory and autonomic fibers in peripheral tissues and high speed of neural transduction allows rapid local and systemic neurogenic modulation of immunity. Peripheral neurons also seem to contribute to immune dysfunction in autoimmune and allergic diseases. Therefore, understanding the coordinated interaction of peripheral neurons with immune cells may advance therapeutic approaches to increase host defense and suppress immunopathology.


Asunto(s)
Sistema Inmunológico/inmunología , Sistema Inmunológico/patología , Inflamación Neurogénica/inmunología , Inflamación Neurogénica/patología , Sistema Nervioso Periférico/inmunología , Sistema Nervioso Periférico/patología , Sistema Nervioso Autónomo/inmunología , Sistema Nervioso Autónomo/patología , Humanos , Nociceptores/patología , Células Receptoras Sensoriales/metabolismo
20.
J Biol Chem ; 283(35): 24136-44, 2008 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-18550530

RESUMEN

Plants, fungi, and animals generate a diverse array of deterrent natural products that induce avoidance behavior in biological adversaries. The largest known chemical family of deterrents are terpenes characterized by reactive alpha,beta-unsaturated dialdehyde moieties, including the drimane sesquiterpenes and other terpene species. Deterrent sesquiterpenes are potent activators of mammalian peripheral chemosensory neurons, causing pain and neurogenic inflammation. Despite their wide-spread synthesis and medicinal use as desensitizing analgesics, their molecular targets remain unknown. Here we show that isovelleral, a noxious fungal sesquiterpene, excites sensory neurons through activation of TPRA1, an ion channel involved in inflammatory pain signaling. TRPA1 is also activated by polygodial, a drimane sesquiterpene synthesized by plants and animals. TRPA1-deficient mice show greatly reduced nocifensive behavior in response to isovelleral, indicating that TRPA1 is the major receptor for deterrent sesquiterpenes in vivo. Isovelleral and polygodial represent the first fungal and animal small molecule agonists of nociceptive transient receptor potential channels.


Asunto(s)
Células Quimiorreceptoras/metabolismo , Dolor/metabolismo , Sesquiterpenos/farmacología , Canales de Potencial de Receptor Transitorio/agonistas , Canales de Potencial de Receptor Transitorio/metabolismo , Analgésicos/farmacología , Animales , Inflamación/metabolismo , Inflamación/fisiopatología , Ratones , Ratones Noqueados , Dolor/inducido químicamente , Dolor/genética , Dolor/fisiopatología , Sesquiterpenos Policíclicos , Canal Catiónico TRPA1 , Canales de Potencial de Receptor Transitorio/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA