Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cancer Treat Res ; 184: 1-39, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36449185

RESUMEN

This study describes cases of individuals who report adverse events following consumption of the most commonly prescribed fluoroquinolone (FQ) antibiotics: ciprofloxacin, levofloxacin, or moxifloxacin. Fluoroquinolone (FQ) antibiotics are some of the most widely prescribed antibiotics in the world. Although these antibiotics have been on the market for more than 20 years, a wide range of serious FQ-associated adverse events first became apparent in 2006 and continued to be recognized for the next 15 years.


Asunto(s)
Antibacterianos , Fluoroquinolonas , Humanos , Fluoroquinolonas/efectos adversos , Antibacterianos/efectos adversos
2.
Expert Opin Drug Saf ; 21(11): 1365-1378, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36384376

RESUMEN

INTRODUCTION: Fluoroquinolone (FQ) antibiotics were approved in 1986 for treatment of urinary tract infections, sinusitis, and bronchitis. Numerous putative FQ-associated adverse events have been recently reported. AREAS COVERED: We review international regulatory agency experience with these FQ-associated toxicities. A 2015 FDA Advisory Committee meeting led regulatory agencies in Canada, Australia, the European Union, New Zealand, and Japan between 2017 and 2021 to evaluate FQ-associated long-term disability and aortic aneurysm/dissections. Regulatory agency guidance in the United States in 2016 warn that FQs should not be used as first-line therapies for urinary tract infections, sinusitis, and bronchitis if other antibiotics are available because of potential long-term and disabling toxicity. Regulatory agencies in European Union countries warn that FQs should not be used to treat mild infections. Product labels in Australia, New Zealand, Japan, and Canada do not have warnings related to FQ-associated disability. Revised product labels and public health advisories in the United States, the European Union, and Japan warn against FQ administration to persons at aortic aneurysm/dissection risks, while product labels and regulatory agency notifications from Canada, Australia, and New Zealand do not include these warnings. EXPERT OPINION: Harmonization of warnings related to FQ-associated disability in particular should be considered.


Asunto(s)
Antibacterianos , Fluoroquinolonas , Humanos , Antibacterianos/efectos adversos , Aneurisma de la Aorta/tratamiento farmacológico , Disección Aórtica , Bronquitis/tratamiento farmacológico , Fluoroquinolonas/efectos adversos , Estados Unidos , Infecciones Urinarias/tratamiento farmacológico
3.
Medicina (Kaunas) ; 57(12)2021 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-34946316

RESUMEN

Background and Objectives: Fluoroquinolones (FQs) are a broad-spectrum class of antibiotics routinely prescribed for common bacterial infections despite recent recommendations to use them only for life-threatening cases. In addition to their antimicrobial properties, FQs act in the central nervous system as GABAA receptor inhibitors, which could potentially affect functionality of the vagus nerve at the forefront of gastrointestinal (GI) tract function. Alterations in neural control of digestion have been shown to be linked to Functional Gastrointestinal Disorders (FGIDs), which are usually diagnosed based on self-reported symptoms. The aim of this study was to assess the incidence of FGIDs following FQ use. Materials and Methods: Self-reports from the FDA Adverse Event Reporting System were analyzed together with ~300 survey responses from a social network derived sample to the Bowel Disease Questionnaire. Results: The results of this study suggested that six different FQs are associated with a wide range of GI symptoms not currently reported in the drugs' labels. The responses from the survey suggested that ~70% of FQ users scored positive for FGID, with no positive correlation between drug type, duration of administration, dosage and frequency of administration. Conclusions: This study showed that GI disorders other than nausea, vomiting and diarrhea are more common than currently reported on the drug labels, and that FGIDs are possibly a common consequence of FQ use even after single use.


Asunto(s)
Infecciones Bacterianas , Enfermedades Gastrointestinales , Antibacterianos/uso terapéutico , Fluoroquinolonas/efectos adversos , Enfermedades Gastrointestinales/inducido químicamente , Enfermedades Gastrointestinales/diagnóstico , Humanos , Encuestas y Cuestionarios
4.
Am J Physiol Gastrointest Liver Physiol ; 317(1): G1-G7, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31042398

RESUMEN

The majority of patients with Parkinson's disease (PD) experience gastrointestinal dysfunction. Recently, we described a nigro-vagal pathway that uses dopaminergic (DA) inputs to the dorsal motor nucleus of the vagus (DMV) and A2 area neurons to modulate gastric motility and tone. This pathway is disrupted in a rodent model of PD. The aim of the present study was to test the hypothesis that brain-stem DA modulation of gastric tone and motility is altered in a rodent model of PD. Male Sprague-Dawley rats received three weekly intraperitoneal injections of paraquat (10 mg/kg) or saline (control). In naive conditions, microinjection of DA into the DMV induced a gastroinhibitory response in 100% of animals. In 19 of 28 PQ-treated animals, however, microinjection of DA into the DVC induced a biphasic response, with an initial increase in gastric tone and motility followed by a profound gastroinhibition. The excitatory response to DA microinjection was attenuated by a combination of DA type 1 (DA1)- and DA2-like receptor antagonists. Conversely, the inhibitory response was reduced by the DA2-like receptor antagonist only. Pretreatment with the α2-adrenoceptor antagonist yohimbine did not modulate the response to DA, thus excluding involvement of the A2 area. At the end of the experiments, induction of the Parkinson phenotype was confirmed by the presence of α-synuclein immunoreactivity in the DMV and substantia nigra pars compacta. These data suggest a maladaptive neural plasticity in brain-stem vagal circuits regulating gastric motility in PQ-treated rats that may be responsible for the gastric dysfunction observed in PD models. NEW & NOTEWORTHY After paraquat treatment and induction of Parkinson's disease, brain-stem dopamine (DA) application induces a biphasic gastric response in the majority of rats, with an initial increase in tone and motility followed by gastroinhibition. The initial increase in gastric tone and motility is mediated via a combined activation of DA type 1 (DA1)- and DA2-like receptors. The inhibitory effects of DA are mediated by DA2-like receptors and are not affected by blockade of adrenergic inputs mediated by α2-adrenoceptors.


Asunto(s)
Tronco Encefálico/metabolismo , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Motilidad Gastrointestinal , Plasticidad Neuronal , Trastornos Parkinsonianos/metabolismo , Estómago/inervación , Nervio Vago/metabolismo , Animales , Tronco Encefálico/fisiopatología , Modelos Animales de Enfermedad , Masculino , Inhibición Neural , Paraquat , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/fisiopatología , Ratas Sprague-Dawley , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Nervio Vago/fisiopatología
5.
J Neurophysiol ; 121(5): 1856-1864, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30917059

RESUMEN

Parkinson's disease (PD) is predominantly idiopathic in origin, and a large body of evidence indicates that gastrointestinal (GI) dysfunctions are a significant comorbid clinical feature; these dysfunctions include dysphagia, nausea, delayed gastric emptying, and severe constipation, all of which occur commonly before the onset of the well-known motor symptoms of PD. Based on a distinct distribution pattern of Lewy bodies (LB) in the enteric nervous system (ENS) and in the preganglionic neurons of the dorsal motor nucleus of the vagus (DMV), and together with the early onset of GI symptoms, it was suggested that idiopathic PD begins in the ENS and spreads to the central nervous system (CNS), reaching the DMV and the substantia nigra pars compacta (SNpc). These two areas are connected by a recently discovered monosynaptic nigro-vagal pathway, which is dysfunctional in rodent models of PD. An alternative hypothesis downplays the role of LB transport through the vagus nerve and proposes that PD pathology is governed by regional or cell-restricted factors as the leading cause of nigral neuronal degeneration. The purpose of this brief review is to summarize the neuronal electrophysiological findings in the SNpc and DMV in PD.


Asunto(s)
Enfermedad de Parkinson/fisiopatología , Sustancia Negra/fisiopatología , Transmisión Sináptica , Animales , Dopamina/metabolismo , Humanos , Enfermedad de Parkinson/metabolismo , Sustancia Negra/metabolismo
6.
Gastroenterology ; 153(6): 1581-1593, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28912019

RESUMEN

BACKGROUND & AIMS: In most patients with Parkinson's disease, gastrointestinal (GI) dysfunctions, such as gastroparesis and constipation, are prodromal to the cardinal motor symptoms of the disease. Sporadic Parkinson's disease has been proposed to develop after ingestion of neurotoxicants that affect the brain-gut axis via the vagus nerve, and then travel to higher centers, compromising the substantia nigra pars compacta (SNpc) and, later, the cerebral cortex. We aimed to identify the pathway that connects the brainstem vagal nuclei and the SNpc, and to determine whether this pathway is compromised in a rat model of Parkinsonism. METHODS: To study this neural pathway in rats, we placed tracers in the dorsal vagal complex or SNpc; brainstem and midbrain were examined for tracer distribution and neuronal neurochemical phenotype. Rats were given injections of paraquat once weekly for 3 weeks to induce features of Parkinsonism, or vehicle (control). Gastric tone and motility were recorded after N-methyl-d-aspartate microinjection in the SNpc and/or optogenetic stimulation of nigro-vagal terminals in the dorsal vagal complex. RESULTS: Stimulation of the SNpc increased gastric tone and motility via activation of dopamine 1 receptors in the dorsal vagal complex. In the paraquat-induced model of Parkinsonism, this nigro-vagal pathway was compromised during the early stages of motor deficit development. CONCLUSIONS: We identified and characterized a nigro-vagal monosynaptic pathway in rats that controls gastric tone and motility. This pathway might be involved in the prodromal gastric dysmotility observed in patients with early-stage Parkinson's disease.


Asunto(s)
Tronco Encefálico/fisiopatología , Vaciamiento Gástrico , Enfermedad de Parkinson Secundaria/fisiopatología , Porción Compacta de la Sustancia Negra/fisiopatología , Estómago/inervación , Nervio Vago/fisiopatología , Animales , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/metabolismo , Modelos Animales de Enfermedad , Vaciamiento Gástrico/efectos de los fármacos , Inmunohistoquímica , Masculino , Actividad Motora , Vías Nerviosas/metabolismo , Vías Nerviosas/fisiopatología , Técnicas de Trazados de Vías Neuroanatómicas , Neurotransmisores/farmacología , Optogenética , Paraquat , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/metabolismo , Porción Compacta de la Sustancia Negra/efectos de los fármacos , Porción Compacta de la Sustancia Negra/metabolismo , Ratas Sprague-Dawley , Receptores de Dopamina D1/metabolismo , Factores de Tiempo , Nervio Vago/efectos de los fármacos , Nervio Vago/metabolismo
7.
J Neurophysiol ; 118(4): 2402-2411, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28747464

RESUMEN

A 2-yr-old boy presented profound developmental delay, failure to thrive, ataxia, hypotonia, and tonic-clonic seizures that caused the death of the patient. Targeted and whole exome sequencing revealed two heterozygous missense variants: a novel mutation in the KCNJ10 gene that encodes for the inward-rectifying K+ channel Kir4.1 and another previously characterized mutation in KCNT1 that encodes for the Na+-activated K+ channel known as Slo2.2 or SLACK. The objectives of this study were to perform the clinical and genetic characterization of the proband and his family and to examine the functional consequence of the Kir4.1 mutation. The mutant and wild-type KCNJ10 constructs were generated and heterologously expressed in Xenopus laevis oocytes, and whole cell K+ currents were measured using the two-electrode voltage-clamp technique. The KCNJ10 mutation c.652C>T resulted in a p.L218F substitution at a highly conserved residue site. Wild-type KCNJ10 expression yielded robust Kir current, whereas currents from oocytes expressing the mutation were reduced, remarkably. Western Blot analysis revealed reduced protein expression by the mutation. Kir5.1 subunits display selective heteromultimerization with Kir4.1 constituting channels with unique kinetics. The effect of the mutation on Kir4.1/5.1 channel activity was twofold: a reduction in current amplitudes and an increase in the pH-dependent inhibition. We thus report a novel loss-of-function mutation in Kir4.1 found in a patient with a coexisting mutation in SLACK channels that results in a fatal disease.NEW & NOTEWORTHY We present and characterize a novel mutation in KCNJ10 Unlike previously reported EAST/SeSAME patients, our patient was heterozygous, and contrary to previous studies, mimicking the heterozygous state by coexpression resulted in loss of channel function. We report in the same patient co-occurrence of a KCNT1 mutation resulting in a more severe phenotype. This study provides new insights into the phenotypic spectrum and to the genotype-phenotype correlations associated with EAST/SeSAME and MMFSI.


Asunto(s)
Discapacidades del Desarrollo/genética , Mutación con Pérdida de Función , Mutación Missense , Proteínas del Tejido Nervioso/genética , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio/genética , Convulsiones/genética , Animales , Discapacidades del Desarrollo/patología , Heterocigoto , Humanos , Lactante , Masculino , Proteínas del Tejido Nervioso/metabolismo , Canales de Potasio/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Canales de potasio activados por Sodio , Convulsiones/patología , Síndrome , Xenopus
8.
Sci Rep ; 7(1): 4583, 2017 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-28676720

RESUMEN

Channelopathy mutations prove informative on disease causing mechanisms and channel gating dynamics. We have identified a novel heterozygous mutation in the KCNA1 gene of a young proband displaying typical signs and symptoms of Episodic Ataxia type 1 (EA1). This mutation is in the S4 helix of the voltage-sensing domain and results in the substitution of the highly conserved phenylalanine 303 by valine (p.F303V). The contributions of F303 towards K+ channel voltage gating are unclear and here have been assessed biophysically and by performing structural analysis using rat Kv1.2 coordinates. We observed significant positive shifts of voltage-dependence, changes in the activation, deactivation and slow inactivation kinetics, reduced window currents, and decreased current amplitudes of both Kv1.1 and Kv1.1/1.2 channels. Structural analysis revealed altered interactions between F303V and L339 and I335 of the S5 helix of a neighboring subunit. The substitution of an aromatic phenylalanine with an aliphatic valine within the voltage-sensor destabilizes the open state of the channel. Thus, F303 fine-tunes the Kv1.1 gating properties and contributes to the interactions between the S4 segment and neighboring alpha helices. The resulting channel's loss of function validates the clinical relevance of the mutation for EA1 pathogenesis.


Asunto(s)
Ataxia/genética , Ataxia/metabolismo , Canalopatías/genética , Canalopatías/metabolismo , Activación del Canal Iónico/genética , Canal de Potasio Kv.1.1/genética , Canal de Potasio Kv.1.1/metabolismo , Mutación , Alelos , Secuencia de Aminoácidos , Ataxia/diagnóstico , Canalopatías/diagnóstico , Secuencia Conservada , Femenino , Genotipo , Humanos , Canal de Potasio Kv.1.1/química , Masculino , Modelos Moleculares , Linaje , Fenilalanina/genética , Conformación Proteica , Evaluación de Síntomas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...