Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Int J Mol Sci ; 23(14)2022 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-35887364

RESUMEN

Breakthrough cancer pain (BTcP) refers to a sudden and transient exacerbation of pain, which develops in patients treated with opioid analgesics. Fast-onset analgesia is required for the treatment of BTcP. Light-activated drugs offer a novel potential strategy for the rapid control of pain without the typical adverse effects of systemic analgesic drugs. mGlu5 metabotropic glutamate receptor antagonists display potent analgesic activity, and light-induced activation of one of these compounds (JF-NP-26) in the thalamus was found to induce analgesia in models of inflammatory and neuropathic pain. We used an established mouse model of BTcP based on the injection of cancer cells into the femur, followed, 16 days later, by systemic administration of morphine. BTcP was induced by injection of endothelin-1 (ET-1) into the tumor, 20 min after morphine administration. Mice were implanted with optic fibers delivering light in the visible spectrum (405 nm) in the thalamus or prelimbic cortex to locally activate systemically injected JF-NP-26. Light delivery in the thalamus caused rapid and substantial analgesia, and this effect was specific because light delivery in the prelimbic cortex did not relieve BTcP. This finding lays the groundwork for the use of optopharmacology in the treatment of BTcP.


Asunto(s)
Analgesia , Dolor Irruptivo , Dolor en Cáncer , Neoplasias , Receptores de Glutamato Metabotrópico , Analgesia/efectos adversos , Analgésicos/farmacología , Analgésicos/uso terapéutico , Analgésicos Opioides/efectos adversos , Animales , Dolor Irruptivo/tratamiento farmacológico , Dolor Irruptivo/etiología , Dolor en Cáncer/tratamiento farmacológico , Dolor en Cáncer/etiología , Modelos Animales de Enfermedad , Ratones , Morfina/farmacología , Morfina/uso terapéutico , Neoplasias/tratamiento farmacológico , Dimensión del Dolor , Tálamo
2.
Clin Case Rep ; 9(11): e04972, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34804526

RESUMEN

Systemic mastocytosis with associated primitive myelofibrosis is a rare and complex disease with a difficult therapeutic management. The release of several inflammation mediators can trigger acute cardiovascular events.

3.
Transl Psychiatry ; 11(1): 109, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33597513

RESUMEN

mGlu5 metabotropic glutamate receptors are highly functional in the early postnatal life, and regulate developmental plasticity of parvalbumin-positive (PV+) interneurons in the cerebral cortex. PV+ cells are enwrapped by perineuronal nets (PNNs) at the closure of critical windows of cortical plasticity. Changes in PNNs have been associated with neurodevelopmental disorders. We found that the number of Wisteria Fluoribunda Agglutinin (WFA)+ PNNs and the density of WFA+/PV+ cells were largely increased in the somatosensory cortex of mGlu5-/- mice at PND16. An increased WFA+ PNN density was also observed after pharmacological blockade of mGlu5 receptors in the first two postnatal weeks. The number of WFA+ PNNs in mGlu5-/- mice was close to a plateau at PND16, whereas continued to increase in wild-type mice, and there was no difference between the two genotypes at PND21 and PND60. mGlu5-/- mice at PND16 showed increases in the transcripts of genes involved in PNN formation and a reduced expression and activity of type-9 matrix metalloproteinase in the somatosensory cortex suggesting that mGlu5 receptors control both PNN formation and degradation. Finally, unilateral whisker stimulation from PND9 to PND16 enhanced WFA+ PNN density in the contralateral somatosensory cortex only in mGlu5+/+ mice, whereas whisker trimming from PND9 to PND16 reduced WFA+ PNN density exclusively in mGlu5-/- mice, suggesting that mGlu5 receptors shape the PNN response to sensory experience. These findings disclose a novel undescribed mechanism of PNN regulation, and lay the groundwork for the study of mGlu5 receptors and PNNs in neurodevelopmental disorders.


Asunto(s)
Receptores de Glutamato Metabotrópico , Corteza Somatosensorial , Animales , Matriz Extracelular/metabolismo , Interneuronas/metabolismo , Ratones , Parvalbúminas/metabolismo
4.
Front Neurol ; 9: 1062, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30564191

RESUMEN

Alzheimer's disease, the most common form of dementia, is marked by progressive cognitive and functional impairment believed to reflect synaptic and neuronal loss. Recent preclinical data suggests that lipopolysaccharide (LPS)-activated microglia may contribute to the elimination of viable neurons and synapses by promoting a neurotoxic astrocytic phenotype, defined as A1. The innate immune cells, including microglia and astrocytes, can either facilitate or inhibit neuroinflammation in response to peripherally applied inflammatory stimuli, such as LPS. Depending on previous antigen encounters, these cells can assume activated (trained) or silenced (tolerized) phenotypes, augmenting or lowering inflammation. Iron, reactive oxygen species (ROS), and LPS, the cell wall component of gram-negative bacteria, are microglial activators, but only the latter can trigger immune tolerization. In Alzheimer's disease, tolerization may be impaired as elevated LPS levels, reported in this condition, fail to lower neuroinflammation. Iron is closely linked to immunity as it plays a key role in immune cells proliferation and maturation, but it is also indispensable to pathogens and malignancies which compete for its capture. Danger signals, including LPS, induce intracellular iron sequestration in innate immune cells to withhold it from pathogens. However, excess cytosolic iron increases the risk of inflammasomes' activation, microglial training and neuroinflammation. Moreover, it was suggested that free iron can awaken the dormant central nervous system (CNS) LPS-shedding microbes, engendering prolonged neuroinflammation that may override immune tolerization, triggering autoimmunity. In this review, we focus on iron-related innate immune pathology in Alzheimer's disease and discuss potential immunotherapeutic agents for microglial de-escalation along with possible delivery vehicles for these compounds.

5.
Sci Rep ; 8(1): 13361, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30190524

RESUMEN

In cerebellar Purkinje cells (PCs) type-1 metabotropic glutamate (mGlu1) receptors play a key role in motor learning and drive the refinement of synaptic innervation during postnatal development. The cognate mGlu5 receptor is absent in mature PCs and shows low expression levels in the adult cerebellar cortex. Here we found that mGlu5 receptors were heavily expressed by PCs in the early postnatal life, when mGlu1α receptors were barely detectable. The developmental decline of mGlu5 receptors coincided with the appearance of mGlu1α receptors in PCs, and both processes were associated with specular changes in CpG methylation in the corresponding gene promoters. It was the mGlu1 receptor that drove the elimination of mGlu5 receptors from PCs, as shown by data obtained with conditional mGlu1α receptor knockout mice and with targeted pharmacological treatments during critical developmental time windows. The suppressing activity of mGlu1 receptors on mGlu5 receptor was maintained in mature PCs, suggesting that expression of mGlu1α and mGlu5 receptors is mutually exclusive in PCs. These findings add complexity to the the finely tuned mechanisms that regulate PC biology during development and in the adult life and lay the groundwork for an in-depth analysis of the role played by mGlu5 receptors in PC maturation.


Asunto(s)
Regulación hacia Abajo , Epigénesis Genética , Células de Purkinje/metabolismo , Receptores AMPA/metabolismo , Receptores de Ácido Kaínico/biosíntesis , Sinapsis/metabolismo , Animales , Islas de CpG , Metilación de ADN , Masculino , Ratones , Ratones Noqueados , Células de Purkinje/citología , Receptores AMPA/genética , Receptores de Ácido Kaínico/genética , Sinapsis/genética
6.
Curr Neuropharmacol ; 15(6): 918-925, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28290248

RESUMEN

BACKGROUND: Spike-wave discharges, underlying absence seizures, are generated within a cortico-thalamo-cortical network that involves the somatosensory cortex, the reticular thalamic nucleus, and the ventrobasal thalamic nuclei. Activation of T-type voltage-sensitive calcium channels (VSCCs) contributes to the pathological oscillatory activity of this network, and some of the first-line drugs used in the treatment of absence epilepsy inhibit T-type calcium channels. The α2δ subunit is a component of high voltage-activated VSCCs (i.e., L-, N-, P/Q-, and R channels) and studies carried out in heterologous expression systems suggest that it may also associate with T channels. The α2δ subunit is also targeted by thrombospondins, which regulate synaptogenesis in the central nervous system. OBJECTIVE: To discuss the potential role for the thrombospondin/α2δ axis in the pathophysiology of absence epilepsy. METHODS: We searched PubMed articles for the terms "absence epilepsy", "T-type voltage-sensitive calcium channels", "α2δ subunit", "ducky mice", "pregabalin", "gabapentin", "thrombospondins", and included papers focusing this Review's scope. RESULTS: We moved from the evidence that mice lacking the α2δ-2 subunit show absence seizures and α 2δ ligands (gabapentin and pregabalin) are detrimental in the treatment of absence epilepsy. This suggests that α2δ may be protective against absence epilepsy via a mechanism that does not involve T channels. We discuss the interaction between thrombospondins and α2δ and its potential relevance in the regulation of excitatory synaptic formation in the cortico-thalamo-cortical network. CONCLUSION: We speculate on the possibility that the thrombospondin/α2 δ axis is critical for the correct functioning of the cortico-thalamo-cortical network, and that abnormalities in this axis may play a role in the pathophysiology of absence epilepsy.


Asunto(s)
Canales de Calcio/metabolismo , Epilepsia Tipo Ausencia/metabolismo , Animales , Epilepsia Tipo Ausencia/tratamiento farmacológico , Humanos , Trombospondinas/metabolismo
7.
Mol Pain ; 13: 1744806917697009, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28326943

RESUMEN

Background L-acetylcarnitine, a drug marketed for the treatment of chronic pain, causes analgesia by epigenetically up-regulating type-2 metabotropic glutamate (mGlu2) receptors in the spinal cord. Because the epigenetic mechanisms are typically long-lasting, we hypothesized that analgesia could outlast the duration of L-acetylcarnitine treatment in models of inflammatory and neuropathic pain. Results A seven-day treatment with L-acetylcarnitine (100 mg/kg, once a day, i.p.) produced an antiallodynic effect in the complete Freund adjuvant mouse model of chronic inflammatory pain. L-Acetylcarnitine-induced analgesia persisted for at least 14 days after drug withdrawal. In contrast, the analgesic effect of pregabalin, amitryptiline, ceftriaxone, and N-acetylcysteine disappeared seven days after drug withdrawal. L-acetylcarnitine treatment enhanced mGlu2/3 receptor protein levels in the dorsal region of the spinal cord. This effect also persisted for two weeks after drug withdrawal and was associated with increased levels of acetylated histone H3 bound to the Grm2 gene promoter in the dorsal root ganglia. A long-lasting analgesic effect of L-acetylcarnitine was also observed in mice subjected to chronic constriction injury of the sciatic nerve. In these animals, a 14-day treatment with pregabalin, amitryptiline, tramadol, or L-acetylcarnitine produced a significant antiallodynic effect, with pregabalin displaying the greatest efficacy. In mice treated with pregabalin, tramadol or L-acetylcarnitine the analgesic effect was still visible 15 days after the end of drug treatment. However, only in mice treated with L-acetylcarnitine analgesia persisted 37 days after drug withdrawal. This effect was associated with an increase in mGlu2/3 receptor protein levels in the dorsal horns of the spinal cord. Conclusions Our findings suggest that L-acetylcarnitine has the unique property to cause a long-lasting analgesic effect that might reduce relapses in patients suffering from chronic pain.


Asunto(s)
Acetilcarnitina/farmacología , Acetilcarnitina/uso terapéutico , Epigénesis Genética/efectos de los fármacos , Inflamación/tratamiento farmacológico , Neuralgia/tratamiento farmacológico , Amitriptilina/uso terapéutico , Analgésicos/farmacología , Analgésicos/uso terapéutico , Animales , Enfermedad Crónica , Modelos Animales de Enfermedad , Adyuvante de Freund/efectos adversos , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/etiología , Inflamación/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Manejo del Dolor , Pregabalina/uso terapéutico , Receptores de Glutamato Metabotrópico/metabolismo , Factores de Tiempo , Tramadol/uso terapéutico
8.
Neuropharmacology ; 115: 51-59, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-27498071

RESUMEN

The neuronal K+/Cl- symporter, KCC2, shapes synaptic responses mediated by Cl--permeant GABAA receptors. Moving from the evidence that excitatory neurotransmission drives changes in KCC2 expression in cerebellar neurons, we studied the regulation of KCC2 expression by group-I metabotropic glutamate (mGlu) receptors in the cerebellum of adult mice. Mice lacking mGlu5 receptors showed a large reduction in cerebellar KCC2 protein levels and a loss of KCC2 immunoreactivity in Purkinje cells. Similar changes were seen in mice treated with the mGlu5 receptor antagonist, MPEP, whereas treatment with the mGlu5 receptor positive allosteric modulator (PAM), VU0360172, increased KCC2 expression. In contrast, pharmacological inhibition of mGlu1 receptors with JNJ16259685 enhanced cerebellar KCC2 protein levels and KCC2 immunoreactivity in Purkinje cells, whereas treatment with the mGlu1 receptor PAM, RO0711401, reduced KCC2 expression. To examine whether the reduction in KCC2 expression caused by the absence or the inhibition of mGlu5 receptors could affect GABAergic transmission, we performed electrophysiological and behavioral studies. Recording of extracellular action potentials in Purkinje cells showed that the inhibitory effect of the GABAA receptor agonist, muscimol, was lost in cerebellar slices prepared from mGlu5-/- mice or from mice treated systemically with MPEP, in line with the reduction in KCC2 expression. Similarly, motor impairment caused by the GABAA receptor PAM, diazepam, was attenuated in mice pre-treated with MPEP. These findings disclose a novel function of mGlu5 receptors in the cerebellum and suggest that mGlu5 receptor ligands might influence GABAergic transmission in the cerebellum and affect motor responses to GABA-mimetic drugs. This article is part of the Special Issue entitled 'Metabotropic Glutamate Receptors, 5 years on'.


Asunto(s)
Cerebelo/metabolismo , Células de Purkinje/metabolismo , Receptores de Glutamato Metabotrópico/fisiología , Simportadores/biosíntesis , Potenciales de Acción/fisiología , Animales , Cerebelo/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/farmacología , Expresión Génica , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Niacinamida/análogos & derivados , Niacinamida/farmacología , Células de Purkinje/efectos de los fármacos , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Simportadores/genética , Cotransportadores de K Cl
9.
Ann Ital Chir ; 872016 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-27830670

RESUMEN

OBJECTIVE: Sinonasal adenocarcinoma is a tumor typically associated with exposure to occupational carcinogens. The International Agency for Research on Cancer (IARC) published several data in order to classify carcinogenic power of physical-chemical agents as far as sinonasal cancer is concerned. MATERIALS AND METHODS: We report a clinical case of sinonasal adenocarcinoma observed in an 84 years old patient, without clinical history of past exposure to carcinogens, smoke and alcohol. RESULTS AND CONCLUSIONS: A sinonasal adenocarcinoma in a patient without risk factors is extremely rare. It is very important to recognize this cancer at an early stage in order to give better survival rates to the patients. KEY WORDS: Carcinogens, Intestinal-type adenocarcinoma, Sinonasal cancer.


Asunto(s)
Adenocarcinoma , Neoplasias de los Senos Paranasales , Adenocarcinoma/diagnóstico , Adenocarcinoma/terapia , Anciano de 80 o más Años , Humanos , Masculino , Neoplasias de los Senos Paranasales/diagnóstico , Neoplasias de los Senos Paranasales/terapia , Factores de Riesgo
10.
Sci Rep ; 5: 17799, 2015 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-26643205

RESUMEN

The kynurenine pathway of tryptophan metabolism has been implicated in the pathophysiology of psychiatric disorders, including schizophrenia. We report here that the kynurenine metabolite, xanturenic acid (XA), interacts with, and activates mGlu2 and mGlu3 metabotropic glutamate receptors in heterologous expression systems. However, the molecular nature of this interaction is unknown, and our data cannot exclude that XA acts primarily on other targets, such as the vesicular glutamate transporter, in the CNS. Systemic administration of XA in mice produced antipsychotic-like effects in the MK-801-induced model of hyperactivity. This effect required the presence of mGlu2 receptors and was abrogated by the preferential mGlu2/3 receptor antagonist, LY341495. Because the mGlu2 receptor is a potential drug target in the treatment of schizophrenia, we decided to measure serum levels of XA and other kynurenine metabolites in patients affected by schizophrenia. Serum XA levels were largely reduced in a large cohort of patients affected by schizophrenia, and, in patients with first-episode schizophrenia, levels remained low after 12 months of antipsychotic medication. As opposed to other kynurenine metabolites, XA levels were also significantly reduced in first-degree relatives of patients affected by schizophrenia. We suggest that lowered serum XA levels might represent a novel trait marker for schizophrenia.


Asunto(s)
Carácter Cuantitativo Heredable , Receptores de Glutamato Metabotrópico/agonistas , Esquizofrenia/diagnóstico , Esquizofrenia/metabolismo , Xanturenatos/metabolismo , Adulto , Anciano , Animales , Biomarcadores , Encéfalo/metabolismo , Estudios de Casos y Controles , Femenino , Células HEK293 , Humanos , Quinurenina/metabolismo , Masculino , Metabolómica/métodos , Ratones , Persona de Mediana Edad , Unión Proteica , Esquizofrenia/sangre , Transducción de Señal , Membranas Sinápticas/metabolismo , Xanturenatos/sangre , Adulto Joven
11.
Front Mol Biosci ; 2: 59, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26579527

RESUMEN

In elderly population sepsis is one of the leading causes of intensive care unit (ICU) admissions in the United States. Sepsis-induced delirium (SID) is the most frequent cause of delirium in ICU (Martin et al., 2010). Together delirium and SID represent under-recognized public health problems which place an increasing financial burden on the US health care system, currently estimated at 143-152 billion dollars per year (Leslie et al., 2008). The interest in SID was recently reignited as it was demonstrated that, contrary to prior beliefs, cognitive deficits induced by this condition may be irreversible and lead to dementia (Pandharipande et al., 2013; Brummel et al., 2014). Conversely, it is construed that diagnosing SID early or mitigating its full blown manifestations may preempt geriatric cognitive disorders. Biological markers specific for sepsis and SID would facilitate the development of potential therapies, monitor the disease process and at the same time enable elderly individuals to make better informed decisions regarding surgeries which may pose the risk of complications, including sepsis and delirium. This article proposes a battery of peripheral blood markers to be used for diagnostic and prognostic purposes in sepsis and SID. Though each individual marker may not be specific enough, we believe that together as a battery they may achieve the necessary accuracy to answer two important questions: who may be vulnerable to the development of sepsis, and who may develop SID and irreversible cognitive deficits following sepsis?

12.
Front Cell Neurosci ; 9: 171, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26029046

RESUMEN

Delirium is an acute state marked by disturbances in cognition, attention, memory, perception, and sleep-wake cycle which is common in elderly. Others have shown an association between delirium and increased mortality, length of hospitalization, cost, and discharge to extended stay facilities. Until recently it was not known that after an episode of delirium in elderly, there is a 63% probability of developing dementia at 48 months compared to 8% in patients without delirium. Currently there are no preventive therapies for delirium, thus elucidation of cellular and molecular underpinnings of this condition may lead to the development of early interventions and thus prevent permanent cognitive damage. In this article we make the case for the role of glia in the pathophysiology of delirium and describe an astrocyte-dependent central and peripheral cholinergic anti-inflammatory shield which may be disabled by astrocytic pathology, leading to neuroinflammation and delirium. We also touch on the role of glia in information processing and neuroimaging.

13.
Neuropharmacology ; 95: 50-8, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25747602

RESUMEN

Neuroadaptive changes involving the indirect pathway of the basal ganglia motor circuit occur in the early phases of parkinsonism. The precise identification of these changes may shed new light into the pathophysiology of parkinsonism and better define the time window of pharmacological intervention. We examined some of these changes in mice challenged with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), or with the dopamine receptor blocker, haloperidol. These two models clearly diverge from Parkinson's disease (PD); however, they allow an accurate time-dependent analysis of neuroadaptive changes occurring in the striatum. Acute haloperidol injection caused a significant increase in the transcripts of mGlu4 receptors, CB1 receptors and preproenkephalin-A at 2 and 24 h, and a reduction in the transcripts of mGlu5 and A2A receptors at 2 h. At least changes in the expression of mGlu4 receptors might be interpreted as compensatory because haloperidol-induced catalepsy was enhanced in mGlu4(-/-) mice. Mice injected with 30 mg/kg of MPTP also showed an increase in the transcripts of mGlu4 receptors, CB1 receptors, and preproenkephalin-A at 3 d, and a reduction of the transcript of A2A receptors at 1 d in the striatum. Genetic deletion of mGlu4 receptors altered the functional response to MPTP, assessed by counting c-Fos(+) neurons in the external globus pallidus and ventromedial thalamic nucleus. These findings offer the first evidence that changes in the expression of mGlu4 and mGlu5 receptors occur in acute models of parkinsonisms, and lay the groundwork for the study of these changes in models that better recapitulate the temporal profile of nigrostriatal dysfunction associated with PD.


Asunto(s)
Cuerpo Estriado/metabolismo , Trastornos Parkinsonianos/metabolismo , Receptor del Glutamato Metabotropico 5/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Enfermedad Aguda , Animales , Catalepsia/metabolismo , Catalepsia/patología , Encefalinas/metabolismo , Haloperidol , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología , Trastornos Parkinsonianos/patología , Precursores de Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptor Cannabinoide CB1/metabolismo , Receptores de Adenosina A2/metabolismo , Receptores de Glutamato Metabotrópico/genética , Factores de Tiempo , Núcleos Talámicos Ventrales/metabolismo
14.
Curr Opin Pharmacol ; 20: 89-94, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25506748

RESUMEN

The question in the title: 'what's new?' has two facets. First, are 'clinical' expectations met with success? Second, is the number of CNS disorders targeted by mGlu drugs still increasing? The answer to the first question is 'no', because development program with promising drugs in the treatment of schizophrenia, Parkinson's disease, and Fragile X syndrome have been discontinued. Nonetheless, we continue to be optimistic because there is still the concrete hope that some of these drugs are beneficial in targeted subpopulations of patients. The answer to the second question is 'yes', because mGlu ligands are promising targets for 'new' disorders such as type-1 spinocerebellar ataxia and absence epilepsy. In addition, the increasing availability of pharmacological tools may push mGlu7 and mGlu8 receptors into the clinical scenario. After almost 30 years from their discovery, mGlu receptors are still alive.


Asunto(s)
Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Diseño de Fármacos , Receptores de Glutamato Metabotrópico/efectos de los fármacos , Animales , Enfermedades del Sistema Nervioso Central/fisiopatología , Epilepsia Tipo Ausencia/tratamiento farmacológico , Epilepsia Tipo Ausencia/fisiopatología , Humanos , Ligandos , Terapia Molecular Dirigida , Receptores de Glutamato Metabotrópico/metabolismo , Ataxias Espinocerebelosas/tratamiento farmacológico , Ataxias Espinocerebelosas/fisiopatología
16.
Mol Brain ; 6: 48, 2013 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-24252411

RESUMEN

BACKGROUND: Spinocerebellar ataxia type 1 (SCA1) is a genetic disorder characterized by severe ataxia associated with progressive loss of cerebellar Purkinje cells. The mGlu1 metabotropic glutamate receptor plays a key role in mechanisms of activity-dependent synaptic plasticity in the cerebellum, and its dysfunction is linked to the pathophysiology of motor symptoms associated with SCA1. We used SCA1 heterozygous transgenic mice (Q154/Q2) as a model for testing the hypothesis that drugs that enhance mGlu1 receptor function may be good candidates for the medical treatment of SCA1. RESULTS: Symptomatic 30-week old SCA1 mice showed reduced mGlu1 receptor mRNA and protein levels in the cerebellum. Interestingly, these mice also showed an intense expression of mGlu5 receptors in cerebellar Purkinje cells, which normally lack these receptors. Systemic treatment of SCA1 mice with the mGlu1 receptor positive allosteric modulator (PAM), Ro0711401 (10 mg/kg, s.c.), caused a prolonged improvement of motor performance on the rotarod and the paw-print tests. A single injection of Ro0711401 improved motor symptoms for several days, and no tolerance developed to the drug. In contrast, the mGlu5 receptor PAM, VU0360172 (10 mg/kg, s.c.), caused only a short-lasting improvement of motor symptoms, whereas the mGlu1 receptor antagonist, JNJ16259685 (2.5 mg/kg, i.p.), further impaired motor performance in SCA1 mice. The prolonged symptomatic benefit caused by Ro0711401 outlasted the time of drug clearance from the cerebellum, and was associated with neuroadaptive changes in the cerebellum, such as a striking reduction of the ectopically expressed mGlu5 receptors in Purkinje cells, increases in levels of total and Ser880-phosphorylated GluA2 subunit of AMPA receptors, and changes in the length of spines in the distal dendrites of Purkinje cells. CONCLUSIONS: These data demonstrate that pharmacological enhancement of mGlu1 receptors causes a robust and sustained motor improvement in SCA1 mice, and lay the groundwork for the development of mGlu1 receptor PAMs as novel "cerebellum-specific", effective, and safe symptomatic drugs for the treatment of SCA1 in humans.


Asunto(s)
Amidas/farmacología , Receptores de Glutamato Metabotrópico/metabolismo , Ataxias Espinocerebelosas/metabolismo , Ataxias Espinocerebelosas/patología , Animales , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Cerebelo/patología , Cromatografía Líquida de Alta Presión , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Espectrometría de Masas , Ratones , Actividad Motora/efectos de los fármacos , Niacinamida/análogos & derivados , Niacinamida/farmacología , Células de Purkinje/efectos de los fármacos , Células de Purkinje/metabolismo , Células de Purkinje/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor del Glutamato Metabotropico 5/genética , Receptor del Glutamato Metabotropico 5/metabolismo , Receptores de Glutamato Metabotrópico/genética , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/fisiopatología
17.
J Pain ; 14(6): 604-12, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23725677

RESUMEN

UNLABELLED: Repeated injections of the antibiotic ceftriaxone cause analgesia in rodents by upregulating the glutamate transporter, GLT-1. No evidence is available in humans. We studied the effect of a single intravenous administration of ceftriaxone in patients undergoing decompressive surgery of the median or ulnar nerves. Forty-five patients were randomized to receive saline, ceftriaxone (2 g), or cefazolin (2 g), 1 hour before surgery. Cefazolin, which is structurally related to ceftriaxone, was used as a negative control. Pain thresholds were measured 10 minutes before drug injections and then 4 to 6 hours after surgery. Ceftriaxone caused analgesia in all patients, whereas cefazolin was inactive. We also performed animal studies to examine whether a single dose of ceftriaxone was sufficient to induce analgesia. A single intraperitoneal injection of ceftriaxone (200 mg/kg), but not cefazoline (200 mg/kg), caused analgesia in mouse models of inflammatory or postsurgical pain, and upregulated GLT-1 in the spinal cord. Ceftriaxone-induced analgesia was additive to that produced by blockade of mGlu5 receptors, which are activated by extrasynaptic glutamate. These data indicate that a single dose of ceftriaxone causes analgesia in humans and mice and suggest that ceftriaxone should be used for preoperative antimicrobial prophylaxis when a fast relief of pain is desired. PERSPECTIVE: The study reports for the first time that a single preoperative dose of ceftriaxone causes analgesia in humans. A single dose of ceftriaxone could also relieve inflammatory and postsurgical pain and upregulate GLT-1 expression in mice. Ceftriaxone should be preferred to other antibiotics for antimicrobial prophylaxis to reduce postoperative pain.


Asunto(s)
Analgésicos/uso terapéutico , Ceftriaxona/uso terapéutico , Dolor Postoperatorio/tratamiento farmacológico , Dolor Postoperatorio/prevención & control , Adulto , Anciano , Anciano de 80 o más Años , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Cefazolina/uso terapéutico , Descompresión Quirúrgica/efectos adversos , Modelos Animales de Enfermedad , Método Doble Ciego , Transportador 2 de Aminoácidos Excitadores/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Actividad Motora/efectos de los fármacos , Dimensión del Dolor , Dolor Postoperatorio/etiología , Enfermedades del Sistema Nervioso Periférico/cirugía , Prueba de Desempeño de Rotación con Aceleración Constante , Factores de Tiempo
18.
PLoS One ; 7(9): e44025, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23028485

RESUMEN

We examined the role of endogenous dopamine (DA) in regulating the number of intrinsic tyrosine hydroxylase-positive (TH(+)) striatal neurons using mice at postnatal day (PND) 4 to 8, a period that corresponds to the developmental peak in the number of these neurons. We adopted the strategy of depleting endogenous DA by a 2-day treatment with α-methyl-p-tyrosine (αMpT, 150 mg/kg, i.p.). This treatment markedly increased the number of striatal TH(+) neurons, assessed by stereological counting, and the increase was highly correlated to the extent of DA loss. Interestingly, TH(+) neurons were found closer to the clusters of DA fibers after DA depletion, indicating that the concentration gradient of extracellular DA critically regulates the distribution of striatal TH(+) neurons. A single i.p. injection of the D1 receptor antagonist, SCH23390 (0.1 mg/kg), the D2/D3 receptor antagonist, raclopride (0.1 mg/kg), or the D4 receptor antagonist, L-745,870 (5 mg/kg) in mice at PND4 also increased the number of TH(+) neurons after 4 days. Treatment with the D1-like receptor agonist SKF38393 (10 mg/kg) or with the D2-like receptor agonist, quinpirole (1 mg/kg) did not change the number of TH(+) neurons. At least the effects of SCH23390 were prevented by a combined treatment with SKF38393. Immunohistochemical analysis indicated that striatal TH(+) neurons expressed D2 and D4 receptors, but not D1 receptors. Moreover, treatment with the α4ß2 receptor antagonist dihydro-ß-erythroidine (DHßE) (3.2 mg/kg) also increased the number of TH(+) neurons. The evidence that DHßE mimicked the action of SCH23390 in increasing the number of TH(+) neurons supports the hypothesis that activation of D1 receptors controls the number of striatal TH(+) neurons by enhancing the release of acetylcholine. These data demonstrate for the first time that endogenous DA negatively regulates the number of striatal TH(+) neurons by direct and indirect mechanisms mediated by multiple DA receptor subtypes.


Asunto(s)
Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Interneuronas/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Animales , Cuerpo Estriado/efectos de los fármacos , Antagonistas de Dopamina/administración & dosificación , Neuronas Dopaminérgicas/efectos de los fármacos , Interneuronas/efectos de los fármacos , Ratones , Fenotipo , Receptores Dopaminérgicos/metabolismo , Receptores de Dopamina D2/metabolismo
19.
Mol Pain ; 8: 77, 2012 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-23088864

RESUMEN

BACKGROUND: Pharmacological activation of type-2 metabotropic glutamate receptors (mGlu2 receptors) causes analgesia in experimental models of inflammatory and neuropathic pain. Presynaptic mGlu2 receptors are activated by the glutamate released from astrocytes by means of the cystine/glutamate antiporter (System x(c)(-) or Sx(c)(-)). We examined the analgesic activity of the Sx(c)(-) activator, N-acetyl-cysteine (NAC), in mice developing inflammatory or neuropathic pain. RESULTS: A single injection of NAC (100 mg/kg, i.p.) reduced nocifensive behavior in the second phase of the formalin test. NAC-induced analgesia was abrogated by the Sxc- inhibitor, sulphasalazine (8 mg/kg, i.p.) or by the mGlu2/3 receptor antagonist, LY341495 (1 mg/kg, i.p.). NAC still caused analgesia in mGlu3(-/-) mice, but was inactive in mGlu2(-/-) mice. In wild-type mice, NAC retained the analgesic activity in the formalin test when injected daily for 7 days, indicating the lack of tolerance. Both single and repeated injections of NAC also caused analgesia in the complete Freund's adjuvant (CFA) model of chronic inflammatory pain, and, again, analgesia was abolished by LY341495. Data obtained in mice developing neuropathic pain in response to chronic constriction injury (CCI) of the sciatic nerve were divergent. In this model, a single injection of NAC caused analgesia that was reversed by LY341495, whereas repeated injections of NAC were ineffective. Thus, tolerance to NAC-induced analgesia developed in the CCI model, but not in models of inflammatory pain. The CFA and CCI models differed with respect to the expression levels of xCT (the catalytic subunit of Sx(c)(-)) and activator of G-protein signaling type-3 (AGS3) in the dorsal portion of the lumbar spinal cord. CFA-treated mice showed no change in either protein, whereas CCI mice showed an ipislateral reduction in xCT levels and a bilateral increase in AGS3 levels in the spinal cord. CONCLUSIONS: These data demonstrate that pharmacological activation of Sxc- causes analgesia by reinforcing the endogenous activation of mGlu2 receptors. NAC has an excellent profile of safety and tolerability when clinically used as a mucolytic agent or in the management of acetaminophen overdose. Thus, our data encourage the use of NAC for the experimental treatment of inflammatory pain in humans.


Asunto(s)
Acetilcisteína/uso terapéutico , Analgésicos/uso terapéutico , Dolor/tratamiento farmacológico , Receptores de Glutamato Metabotrópico/metabolismo , Sistema de Transporte de Aminoácidos y+/genética , Sistema de Transporte de Aminoácidos y+/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Glutamato Metabotrópico/genética
20.
Psychoneuroendocrinology ; 37(10): 1646-58, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22444623

RESUMEN

Epidemiological studies suggest that emotional liability in infancy could be a predictor of anxiety-related disorders in the adulthood. Rats exposed to prenatal restraint stress ("PRS rats") represent a valuable model for the study of the interplay between environmental triggers and neurodevelopment in the pathogenesis of anxious/depressive like behaviours. Repeated episodes of restraint stress were delivered to female Sprague-Dawley rats during pregnancy and male offspring were studied. Ultrasonic vocalization (USV) was assessed in pups under different behavioural paradigms. After weaning, anxiety was measured by conventional tests. Expression of GABA(A) receptor subunits and metabotropic glutamate (mGlu) receptors was assessed by immunoblotting. Plasma leptin levels were measured using a LINCOplex bead assay kit. The offspring of stressed dams emitted more USVs in response to isolation from their mothers and showed a later suppression of USV production when exposed to an unfamiliar male odour, indicating a pronounced anxiety-like profile. Anxiety like behaviour in PRS pups persisted one day after weaning. PRS pups did not show the plasma peak in leptin levels that is otherwise seen at PND14. In addition, PRS pups showed a reduced expression of the γ2 subunit of GABA(A) receptors in the amygdala at PND14 and PND22, an increased expression of mGlu5 receptors in the amygdala at PND22, a reduced expression of mGlu5 receptors in the hippocampus at PND14 and PND22, and a reduced expression of mGlu2/3 receptors in the hippocampus at PND22. These data offer a clear-cut demonstration that the early programming triggered by PRS could be already translated into anxiety-like behaviour during early postnatal life.


Asunto(s)
Ansiedad/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Estrés Psicológico/metabolismo , Vocalización Animal , Amígdala del Cerebelo/metabolismo , Animales , Ansiedad/sangre , Ansiedad/psicología , Femenino , Hipocampo/metabolismo , Leptina/sangre , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/psicología , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/biosíntesis , Receptores de Glutamato Metabotrópico/biosíntesis , Restricción Física
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...