Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Intervalo de año de publicación
1.
Cells ; 12(5)2023 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-36899886

RESUMEN

V-ATPase is an important factor in synaptic vesicle acidification and is implicated in synaptic transmission. Rotation in the extra-membranous V1 sector drives proton transfer through the membrane-embedded multi-subunit V0 sector of the V-ATPase. Intra-vesicular protons are then used to drive neurotransmitter uptake by synaptic vesicles. V0a and V0c, two membrane subunits of the V0 sector, have been shown to interact with SNARE proteins, and their photo-inactivation rapidly impairs synaptic transmission. V0d, a soluble subunit of the V0 sector strongly interacts with its membrane-embedded subunits and is crucial for the canonic proton transfer activity of the V-ATPase. Our investigations show that the loop 1.2 of V0c interacts with complexin, a major partner of the SNARE machinery and that V0d1 binding to V0c inhibits this interaction, as well as V0c association with SNARE complex. The injection of recombinant V0d1 in rat superior cervical ganglion neurons rapidly reduced neurotransmission. In chromaffin cells, V0d1 overexpression and V0c silencing modified in a comparable manner several parameters of unitary exocytotic events. Our data suggest that V0c subunit promotes exocytosis via interactions with complexin and SNAREs and that this activity can be antagonized by exogenous V0d.


Asunto(s)
Proteínas SNARE , ATPasas de Translocación de Protón Vacuolares , Ratas , Animales , Proteínas SNARE/metabolismo , Protones , Vesículas Sinápticas/metabolismo , Fusión de Membrana , ATPasas de Translocación de Protón Vacuolares/metabolismo
2.
Neuron ; 111(2): 202-219.e7, 2023 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-36368315

RESUMEN

In addition to tau and Aß pathologies, inflammation plays an important role in Alzheimer's disease (AD). Variants in APOE and TREM2 increase AD risk. ApoE4 exacerbates tau-linked neurodegeneration and inflammation in P301S tau mice and removal of microglia blocks tau-dependent neurodegeneration. Microglia adopt a heterogeneous population of transcriptomic states in response to pathology, at least some of which are dependent on TREM2. Previously, we reported that knockout (KO) of TREM2 attenuated neurodegeneration in P301S mice that express mouse Apoe. Because of the possible common pathway of ApoE and TREM2 in AD, we tested whether TREM2 KO (T2KO) would block neurodegeneration in P301S Tau mice expressing ApoE4 (TE4), similar to that observed with microglial depletion. Surprisingly, we observed exacerbated neurodegeneration and tau pathology in TE4-T2KO versus TE4 mice, despite decreased TREM2-dependent microgliosis. Our results suggest that tau pathology-dependent microgliosis, that is, TREM2-independent microgliosis, facilitates tau-mediated neurodegeneration in the presence of ApoE4.


Asunto(s)
Enfermedad de Alzheimer , Apolipoproteína E4 , Ratones , Animales , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Enfermedad de Alzheimer/metabolismo , Inflamación/metabolismo , Microglía/metabolismo , Modelos Animales de Enfermedad , Glicoproteínas de Membrana/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo
3.
J Neuroinflammation ; 19(1): 54, 2022 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-35197067

RESUMEN

BACKGROUND: In the field of autoimmune demyelinating diseases, visual impairments have extensively been studied using the experimental autoimmune encephalomyelitis (EAE) mouse model, which is classically induced by immunization with myelin oligodendrocyte glycoprotein peptide (MOG35-55). However, this model does not involve B cells like its human analogs. New antigens have thus been developed to induce a B cell-dependent form of EAE that better mimics human diseases. METHODS: The present study aimed to characterize the visual symptoms of EAE induced with such an antigen called bMOG. After the induction of EAE with bMOG in C57BL/6J mice, visual function changes were studied by electroretinography and optomotor acuity tests. Motor deficits were assessed in parallel with a standard clinical scoring method. Histological examinations and Western blot analyses allowed to follow retinal neuron survival, gliosis, microglia activation, opsin photopigment expression in photoreceptors and optic nerve demyelination. Disease effects on retinal gene expression were established by RNA sequencing. RESULTS: We observed that bMOG EAE mice exhibited persistent loss of visual acuity, despite partial recovery of electroretinogram and motor functions. This loss was likely due to retinal inflammation, gliosis and synaptic impairments, as evidenced by histological and transcriptomic data. Further analysis suggests that the M-cone photoreceptor pathway was also affected. CONCLUSION: Therefore, by documenting visual changes induced by bMOG and showing similarities to those seen in diseases such as multiple sclerosis and neuromyelitis optica, this study offers a new approach to test protective or restorative ophthalmic treatments.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Animales , Electrorretinografía , Encefalomielitis Autoinmune Experimental/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito , Nervio Óptico/patología
4.
Neurobiol Dis ; 161: 105542, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34737043

RESUMEN

BACKGROUND: Vitamin A (VitA), via its active metabolite retinoic acid (RA), is critical for the maintenance of memory function with advancing age. Although its role in Alzheimer's disease (AD) is not well understood, data suggest that impaired brain VitA signaling is associated with the accumulation of ß-amyloid peptides (Aß), and could thus contribute to the onset of AD. METHODS: We evaluated the protective action of a six-month-long dietary VitA-supplementation (20 IU/g), starting at 8 months of age, on the memory and the neuropathology of the 3xTg-AD mouse model of AD (n = 11-14/group; including 4-6 females and 7-8 males). We also measured protein levels of Retinoic Acid Receptor ß (RARß) and Retinoid X Receptor γ (RXRγ) in homogenates from the inferior parietal cortex of 60 participants of the Religious Orders study (ROS) divided in three groups: no cognitive impairment (NCI) (n = 20), mild cognitive impairment (MCI) (n = 20) and AD (n = 20). RESULTS: The VitA-enriched diet preserved spatial memory of 3xTg-AD mice in the Y maze. VitA-supplementation affected hippocampal RXR expression in an opposite way according to sex by tending to increase in males and decrease in females their mRNA expression. VitA-enriched diet also reduced the amount of hippocampal Aß40 and Aß42, as well as the phosphorylation of tau protein at sites Ser396/Ser404 (PHF-1) in males. VitA-supplementation had no effect on tau phosphorylation in females but worsened their hippocampal Aß load. However, the expression of Rxr-ß in the hippocampus was negatively correlated with the amount of both soluble and insoluble Aß in both males and females. Western immunoblotting in the human cortical samples of the ROS study did not reveal differences in RARß levels. However, it evidenced a switch from a 60-kDa-RXRγ to a 55-kDa-RXRγ in AD, correlating with ante mortem cognitive decline and the accumulation of neuritic plaques in the brain cortex. CONCLUSION: Our data suggest that (i) an altered expression of RXRs receptors is a contributor to ß-amyloid pathology in both humans and 3xTg-AD mice, (ii) a chronic exposure of 3xTg-AD mice to a VitA-enriched diet may be protective in males, but not in females.


Asunto(s)
Enfermedad de Alzheimer , Vitamina A , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Dieta , Modelos Animales de Enfermedad , Femenino , Hipocampo/metabolismo , Humanos , Masculino , Ratones , Ratones Transgénicos , Receptores X Retinoide/metabolismo , Proteínas tau/metabolismo
5.
Int J Mol Sci ; 22(21)2021 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-34768774

RESUMEN

We have previously reported that vision decline was not associated with amyloidogenesis processing in aging C57BL/6J wild-type (WT) mice and in a mouse model of Alzheimer's disease, the APPswe/PS1ΔE9 transgenic mouse model (APP/PS1). This conclusion was drawn using middle-aged (10-13 months old) mice. Here, we hypothesized that compared with hippocampal and cortical neurons, the weak amyloidogenic activity of retinal neurons may result in a detectable release of amyloid ß (Aß) only in aged mice, i.e., between 14 and 24 months of age. The aim of the present study was thus to follow potential activity changes in the amyloidogenic and nonamyloidogenic pathways of young (4 months) and old (20-24 months) WT and APP/PS1 mice. Our results showed that in spite of retinal activity loss reported by electroretinogram (ERG) recordings, the level of amyloid beta precursor protein (APP) and its derivatives did not significantly vary in the eyes of old vs. young mice. Strikingly, the ectopic expression of human APPswe in APP/PS1 mice did not allow us to detect Aß monomers at 23 months. In contrast, Aß was observed in hippocampal and cortical tissues at this age but not at 4 months of life. In contrast, optic nerve transection-induced retinal ganglion cell injury significantly affected the level of retinal APP and the secretion of soluble APP alpha in the vitreous. Collectively, these results suggest that the amyloidogenic and nonamyloidogenic pathways are not involved in visual function decline in aging mice. In WT and APP/PS1 mice, it is proposed that retinal neurons do not have the capacity to secrete Aß in contrast with other cortical and hippocampal neurons.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas del Ojo/metabolismo , Retina/metabolismo , Factores de Edad , Animales , Modelos Animales de Enfermedad , Electrorretinografía , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Traumatismos del Nervio Óptico/metabolismo , Retina/diagnóstico por imagen , Células Ganglionares de la Retina/metabolismo
6.
Int J Mol Sci ; 22(21)2021 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-34768981

RESUMEN

We present here a gene therapy approach aimed at preventing the formation of Ca2+-permeable amyloid pore oligomers that are considered as the most neurotoxic structures in both Alzheimer's and Parkinson's diseases. Our study is based on the design of a small peptide inhibitor (AmyP53) that combines the ganglioside recognition properties of the ß-amyloid peptide (Aß, Alzheimer) and α-synuclein (α-syn, Parkinson). As gangliosides mediate the initial binding step of these amyloid proteins to lipid rafts of the brain cell membranes, AmyP53 blocks, at the earliest step, the Ca2+ cascade that leads to neurodegeneration. Using a lentivirus vector, we genetically modified brain cells to express the therapeutic coding sequence of AmyP53 in a secreted form, rendering these cells totally resistant to oligomer formation by either Aß or α-syn. This protection was specific, as control mCherry-transfected cells remained fully sensitive to these oligomers. AmyP53 was secreted at therapeutic concentrations in the supernatant of cultured cells, so that the therapy was effective for both transfected cells and their neighbors. This study is the first to demonstrate that a unique gene therapy approach aimed at preventing the formation of neurotoxic oligomers by targeting brain gangliosides may be considered for the treatment of two major neurodegenerative disorders, Alzheimer's and Parkinson's diseases.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Fragmentos de Péptidos/metabolismo , Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Proteínas Amiloidogénicas/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Calcio/metabolismo , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Gangliósidos/metabolismo , Terapia Genética/métodos , Humanos
7.
Neurobiol Aging ; 95: 214-224, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32858248

RESUMEN

Tau is a microtubule-associated protein involved in Alzheimer's disease. However, little is known on its physiological function in the healthy central nervous system. Here, we observed that the expression of Tau isoforms was modulated by neuronal maturation and visual experience in the mouse retina and in the visual cortex. The visual function of wild-type (WT) and Tau knockout (KO) mice was evaluated using the optokinetic reflex (OKR), an innate visuomotor behavior, and by electroretinography. Visual tests did not reveal functional impairments in young adult and old Tau KO animals. Moreover, monocular deprivation (MD) was used to increase OKR sensitivity, a plasticity phenomenon depending on the visual cortex. MD-induced OKR sensitivity enhancement was significantly stronger in Tau KO than in WT mice suggesting that Tau restricts visual plasticity. In addition, human Tau expression did not affect visual function and plasticity in a mouse tauopathy model, relative to WT controls. Our results unveil a novel function for Tau in the adaptive mechanisms of plasticity operating in the adult brain subjected to sensory experience changes.


Asunto(s)
Envejecimiento/metabolismo , Envejecimiento/fisiología , Plasticidad Neuronal/genética , Corteza Visual/fisiología , Proteínas tau/metabolismo , Proteínas tau/fisiología , Adaptación Fisiológica/genética , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones Noqueados , Retina/metabolismo , Tauopatías/fisiopatología , Corteza Visual/metabolismo
8.
Int J Mol Sci ; 21(11)2020 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-32521826

RESUMEN

In the present study, we hypothesized that the microtubule-associated protein Tau may influence retinal neuron survival and axonal regeneration after optic nerve injury. To test this hypothesis, the density of retinal ganglion cells was evaluated by immunostaining retinal flat-mounts for RNA-binding protein with multiple splicing (RBPMS) two weeks after optic nerve micro-crush lesion in Tau-deprived (Tau knock-out (KO)) and wild-type (WT) mice. Axon growth was determined on longitudinal sections of optic nerves after anterograde tracing. Our results showed that the number of surviving retinal ganglion cells and growing axons did not significantly vary between WT and Tau KO animals. Moreover, sustained activation of the neuronal growth program with ciliary neurotrophic factor (CNTF) resulted in a similar increase in surviving neurons and in growing axons in WT and Tau KO mice. Taken together, our data suggest that Tau does not influence axonal regeneration or neuronal survival.


Asunto(s)
Axones/metabolismo , Eliminación de Gen , Regeneración Nerviosa/genética , Traumatismos del Nervio Óptico/genética , Traumatismos del Nervio Óptico/metabolismo , Células Ganglionares de la Retina/patología , Proteínas tau/genética , Animales , Muerte Celular , Supervivencia Celular , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Ratones , Ratones Noqueados , Traumatismos del Nervio Óptico/patología , Retina/metabolismo , Retina/patología
9.
Cell Death Dis ; 11(2): 101, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-32029703

RESUMEN

N-Methyl-D-aspartate (NMDA)-induced neuronal cell death is involved in a large spectrum of diseases affecting the brain and the retina such as Alzheimer's disease and diabetic retinopathy. Associated neurological impairments may result from the inhibition of neuronal plasticity by Nogo-A. The objective of the current study was to determine the contribution of Nogo-A to NMDA excitotoxicity in the mouse retina. We observed that Nogo-A is upregulated in the mouse vitreous during NMDA-induced inflammation. Intraocular injection of a function-blocking antibody specific to Nogo-A (11C7) was carried out 2 days after NMDA-induced injury. This treatment significantly enhanced visual function recovery in injured animals. Strikingly, the expression of potent pro-inflammatory molecules was downregulated by 11C7, among which TNFα was the most durably decreased cytokine in microglia/macrophages. Additional analyses suggest that TNFα downregulation may stem from cofilin inactivation in microglia/macrophages. 11C7 also limited gliosis presumably via P.Stat3 downregulation. Diabetic retinopathy was associated with increased levels of Nogo-A in the eyes of donors. In summary, our results reveal that Nogo-A-targeting antibody can stimulate visual recovery after retinal injury and that Nogo-A is a potent modulator of excitotoxicity-induced neuroinflammation. These data may be used to design treatments against inflammatory eye diseases.


Asunto(s)
Células Amacrinas/efectos de los fármacos , Antiinflamatorios/farmacología , Anticuerpos Neutralizantes/farmacología , Proteínas Nogo/antagonistas & inhibidores , Células Ganglionares de la Retina/efectos de los fármacos , Retinitis/prevención & control , Visión Ocular/efectos de los fármacos , Anciano , Anciano de 80 o más Años , Células Amacrinas/metabolismo , Células Amacrinas/patología , Animales , Retinopatía Diabética/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , N-Metilaspartato , Plasticidad Neuronal/efectos de los fármacos , Proteínas Nogo/metabolismo , Fosforilación , Recuperación de la Función , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Retinitis/inducido químicamente , Retinitis/metabolismo , Retinitis/fisiopatología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
11.
Front Mol Neurosci ; 11: 293, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30197586

RESUMEN

The implication of the microtubule-associated protein (MAP) Tau in the ocular manifestations of Alzheimer's disease (AD) is elusive due to the lack of relevant animal model. However, signs of AD have been reported in the brain of transgenic mice expressing human Tau (hTau). To assess whether hTau is sufficient to induce AD pathogenesis in the retina as well, in the present study, we compared the retinal structure and function of KO mice deprived of Tau (mTKO) with those of transgenic mice expressing hTau. Our results revealed that hTau is particularly abundant in the inner nuclear layer (INL) cells of the retina. By electroretinogram (ERG) recording, light-induced retinal cell activation was not altered in hTau compared with mTKO littermates. Surprisingly, the ERG response mediated by cone photoreceptor stimulation was even stronger in hTau than in mTKO retinae. Immunofluorescent analysis of retinal sections allowed us to observe thicker inner retina in hTau than in mTKO eyes. By Western Blotting (WB), the upregulation of mTOR that was found in hTau mice may underlie retinal structure and function increases. Taken together, our results not only indicate that hTau expression is not toxic for retinal cells but they also suggest that it may play a positive role in visual physiology. The use of hTau may be envisaged to improve visual recovery in ocular diseases affecting the retinal function such as glaucoma or diabetic retinopathy.

12.
Glia ; 66(10): 2079-2093, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30051920

RESUMEN

Nogo-A is a potent glial-derived inhibitor of axon growth in the injured CNS and acts as a negative regulator of developmental angiogenesis by inhibiting vascular endothelial cell migration. However, its function in pathological angiogenesis has never been studied after ischemic injury in the CNS. Using the mouse model of oxygen-induced retinopathy (OIR) which yields defined zones of retinal ischemia, our goal was to investigate the role of Nogo-A in vascular regeneration. We demonstrate a marked upregulation of the Nogo-A receptor sphingosine 1-phosphate receptor 2 in blood vessels following OIR, while Nogo-A is abundantly expressed in surrounding glial cells. Acute inhibition of Nogo-A with function-blocking antibody 11C7 significantly improved vascular regeneration and consequently prevented pathological pre-retinal angiogenesis. Ultimately, inhibition of Nogo-A led to restoration of retinal function as determined by electrophysiological response of retinal cells to light stimulation. Our data suggest that anti-Nogo-A antibody may protect neuronal cells from ischemic damage by accelerating blood vessel repair in the CNS. Targeting Nogo-A by immunotherapy may improve CNS perfusion after vascular injuries.


Asunto(s)
Isquemia/metabolismo , Neovascularización Fisiológica/fisiología , Proteínas Nogo/metabolismo , Regeneración/fisiología , Enfermedades de la Retina/metabolismo , Vasos Retinianos/metabolismo , Inductores de la Angiogénesis/farmacología , Animales , Modelos Animales de Enfermedad , Isquemia/tratamiento farmacológico , Isquemia/patología , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Neuroglía/patología , Proteínas Nogo/antagonistas & inhibidores , Proteínas Nogo/inmunología , Receptores de Lisoesfingolípidos/metabolismo , Regeneración/efectos de los fármacos , Enfermedades de la Retina/tratamiento farmacológico , Enfermedades de la Retina/patología , Vasos Retinianos/efectos de los fármacos , Vasos Retinianos/patología , Receptores de Esfingosina-1-Fosfato , Visión Ocular/efectos de los fármacos , Visión Ocular/fisiología
13.
Cell Death Dis ; 9(7): 727, 2018 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-29950598

RESUMEN

Myelin-associated proteins such as Nogo-A are major inhibitors of neuronal plasticity that contribute to permanent neurological impairments in the injured CNS. In the present study, we investigated the influence of Nogo-A on visual recovery after retinal injuries in mice. Different doses of N-methyl-D-aspartate (NMDA) were injected in the vitreous of the left eye to induce retinal neuron death. The visual function was monitored using the optokinetic response (OKR) as a behavior test, and electroretinogram (ERG) and local field potential (LFP) recordings allowed to assess changes in retinal and cortical neuron activity, respectively. Longitudinal OKR follow-ups revealed reversible visual deficits after injection of NMDA ≤ 1 nmole in the left eye and concomitant functional improvement in the contralateral visual pathway of the right eye that was let intact. Irreversible OKR loss observed with NMDA ≥ 2 nmol was correlated with massive retinal cell death and important ERG response decline. Strikingly, the OKR mediated by injured and intact eye stimulation was markedly improved in Nogo-A KO mice compared with WT animals, suggesting that the inactivation of Nogo-A promotes visual recovery and plasticity. Moreover, OKR improvement was associated with shorter latency of the N2 wave of Nogo-A KO LFPs relative to WT animals. Strikingly, intravitreal injection of anti-Nogo-A antibody (11C7) in the injured eye exerted positive effects on cortical LFPs. This study presents the intrinsic ability of the visual system to recover from NMDA-induced retinal injury and its limitations. Nogo-A neutralization may promote visual recovery in retinal diseases such as glaucoma.


Asunto(s)
Plasticidad Neuronal , Proteínas Nogo/metabolismo , Recuperación de la Función , Retina/lesiones , Retina/fisiopatología , Visión Ocular/fisiología , Animales , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Neutralizantes/farmacología , Regulación hacia Abajo/efectos de los fármacos , Electrorretinografía , Regulación de la Expresión Génica/efectos de los fármacos , Inyecciones Intravítreas , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , N-Metilaspartato/administración & dosificación , N-Metilaspartato/farmacología , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Recuperación de la Función/efectos de los fármacos , Retina/efectos de los fármacos , Retina/patología , Neuronas Retinianas/efectos de los fármacos , Neuronas Retinianas/metabolismo
14.
Proc Natl Acad Sci U S A ; 114(29): 7719-7724, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28673977

RESUMEN

Autosomal dominant epilepsy with auditory features results from mutations in leucine-rich glioma-inactivated 1 (LGI1), a soluble glycoprotein secreted by neurons. Animal models of LGI1 depletion display spontaneous seizures, however, the function of LGI1 and the mechanisms by which deficiency leads to epilepsy are unknown. We investigated the effects of pure recombinant LGI1 and genetic depletion on intrinsic excitability, in the absence of synaptic input, in hippocampal CA3 neurons, a classical focus for epileptogenesis. Our data indicate that LGI1 is expressed at the axonal initial segment and regulates action potential firing by setting the density of the axonal Kv1.1 channels that underlie dendrotoxin-sensitive D-type potassium current. LGI1 deficiency incurs a >50% down-regulation of the expression of Kv1.1 and Kv1.2 via a posttranscriptional mechanism, resulting in a reduction in the capacity of axonal D-type current to limit glutamate release, thus contributing to epileptogenesis.


Asunto(s)
Axones/metabolismo , Proteínas/metabolismo , Canales de Potasio de la Superfamilia Shaker/metabolismo , Potenciales de Acción , Animales , Venenos Elapídicos/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Canal de Potasio Kv.1.2/metabolismo , Ratones Mutantes , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , Proteínas/genética , Proteínas/farmacología , Ratas Wistar , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología
15.
Ann. hepatol ; 16(3): 430-435, May.-Jun. 2017. tab, graf
Artículo en Inglés | LILACS | ID: biblio-887255

RESUMEN

ABSTRACT Introduction. There is little information on survival rates of patients with primary biliary cholangtis (PBC) in developing countries. This is particularly true in Latin America, where the number of liver transplants performed remains extremely low for patients with advanced liver disease who fulfill criteria for liver transplantation. The goal of this study was to compare survival rate of patients with PBC in developing countries who were treated with ursodeoxycholic acid (UDCA) versus survival of patients who received other treatments (OT) without UDCA, prescribed before the UDCA era. Material and methods. A retrospective study was performed, including records of 78 patients with PBC in the liver unit in a third level referral hospital in Mexico City. Patients were followed for five years from initial diagnosis until death related to liver disease or to the end of the study. Patients received UDCA (15 mg/kg/per day) (n = 41) or OT (n = 37) before introduction of UDCA in Mexico. Results. Response to treatment was higher in the group that received UDCA. In the five years of follow-up, survival rates were significantly higher in the UDCA group than in the OT group. The hazard ratio of death was higher in the OT group vs. UDCA group, HR 8.78 (95% Cl, 2.52-30.61); Mayo Risk Score and gender were independently associated with the risk of death. Conclusions. The study confirms that the use of UDCA in countries with a limited liver transplant program increases survival in comparison to other treatments used before the introduction of UDCA.(AU)


Asunto(s)
Humanos , Ácido Ursodesoxicólico/uso terapéutico , Trasplante de Hígado/efectos adversos , Cirrosis Hepática Biliar/fisiopatología , Tasa de Supervivencia , Estudios Retrospectivos , América Latina
16.
Sci Rep ; 6: 28781, 2016 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-27352802

RESUMEN

Calcium-permeable pores formed by small oligomers of amyloid proteins are the primary pathologic species in Alzheimer's and Parkinson's diseases. However, the molecular mechanisms underlying the assembly of these toxic oligomers in the plasma membrane of brain cells remain unclear. Here we have analyzed and compared the pore-forming capability of a large panel of amyloid proteins including wild-type, variant and truncated forms, as well as synthetic peptides derived from specific domains of Aß1-42 and α-synuclein. We show that amyloid pore formation involves two membrane lipids, ganglioside and cholesterol, that physically interact with amyloid proteins through specific structural motifs. Mutation or deletion of these motifs abolished pore formation. Moreover, α-synuclein (Parkinson) and Aß peptide (Alzheimer) did no longer form Ca(2+)-permeable pores in presence of drugs that target either cholesterol or ganglioside or both membrane lipids. These results indicate that gangliosides and cholesterol cooperate to favor the formation of amyloid pores through a common molecular mechanism that can be jammed at two different steps, suggesting the possibility of a universal therapeutic approach for neurodegenerative diseases. Finally we present the first successful evaluation of such a new therapeutic approach (coined "membrane therapy") targeting amyloid pores formed by Aß1-42 and α-synuclein.


Asunto(s)
Péptidos beta-Amiloides/fisiología , alfa-Sinucleína/fisiología , Secuencia de Aminoácidos , Péptidos beta-Amiloides/química , Calcio/metabolismo , Línea Celular Tumoral , Permeabilidad de la Membrana Celular , Colesterol/fisiología , Ciclodextrinas/farmacología , Gangliósidos/fisiología , Humanos , Fragmentos de Péptidos/farmacología , Dominios Proteicos , alfa-Sinucleína/química
17.
Peptides ; 35(1): 122-30, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22406227

RESUMEN

In the honey bee no allatotropin gene has been found, even though allatotropin stimulates the synthesis of juvenile hormone in this species. We report here that honey bees and other Hymenoptera do have a typical allatotropin gene, although the peptides predicted have a somewhat different structure from that of other insect allatotropins. Polyclonal antisera to honey bee allatotropin reacted with material in the neurohemal organs of the segmental nerves of abdominal ganglia. We were unable to find the allatotropin peptide using mass spectrometry in extracts from these tissues. Thus the expression of this gene in honey bees is less important than in other insect species. We also characterized the leucokinin gene which similarly appears to be very weakly expressed in worker honey bees. Unlike the allatotropin gene, which is conserved within Hymenoptera, the leucokinin gene is much more variable in structure and was not found in ants nor the parasitic wasp Nasonia vitripennis. The absence of significant expression of adipokinetic hormone (AKH) in the honey bee may be due to the existence of a second TATA box in the promotor region of the gene, which explains the production of an mRNA encoding a putative peptide precursor from which no AKH should be released. Such a second TATA box was not found in other Hymenoptera, and may therefore be specific for the two Apis species. It is suggested that functional disintegration of this important metabolic gene became possible in Apis because of the highly evolved social nature of the species.


Asunto(s)
Abejas/genética , Hormonas de Insectos/genética , Proteínas de Insectos/genética , Neuropéptidos/genética , Oligopéptidos/genética , Ácido Pirrolidona Carboxílico/análogos & derivados , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Ganglios de Invertebrados/metabolismo , Himenópteros/genética , Hormonas de Insectos/metabolismo , Proteínas de Insectos/metabolismo , Datos de Secuencia Molecular , Neuropéptidos/metabolismo , Oligopéptidos/metabolismo , Ácido Pirrolidona Carboxílico/metabolismo , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...