Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 202
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Appl Clin Med Phys ; : e14516, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39287608

RESUMO

PURPOSE: The presence of metal implants can produce artifacts and distort Hounsfield units (HU) in patient computed tomography (CT) images. The purpose of this work was to characterize a novel metal artifact reduction (MAR) algorithm for reconstruction of CBCT images obtained by the HyperSight imaging system. METHODS: Three tissue-equivalent phantoms were fitted with materials commonly used in medical applications. The first consisted of a variety of metal samples centered within a solid water block, the second was an Advanced Electron Density phantom with metal rods, and the third consisted of hip prostheses positioned within a water tank. CBCT images of all phantoms were acquired and reconstructed using the MAR and iCBCT Acuros algorithms on the HyperSight system. The signal-to-noise ratio (SNR), artifact index (AI), structural similarity index measure (SSIM), peak signal-to-noise ratio (PSNR), and mean-square error (MSE) were computed to assess the image quality in comparison to artifact-free reference images. The mean HU at various VOI positions around the cavity was calculated to evaluate the artifact dependence on distance and angle from the center of the cavity. The artifact volume of the phantom (excluding the cavity) was estimated by summing the volume of all voxels with HU values outside the 5th and 95th percentiles of the phantom CBCT with no artifact. RESULTS: The SNR, AI, SSIM, PSNR, and MSE metrics demonstrated significantly higher similarity to baseline when using MAR compared to iCBCT Acuros for all high-density materials, except for aluminum. Mean HU returned to expected solid water background at a shorter distance from metal sample in the MAR images, and the standard deviation remained lower for the MAR images at all distances from the insert. The artifact volume decreased using the novel MAR algorithm for all metal samples excluding aluminum (p < 0.001) and all hip prostheses (p < 0.05). CONCLUSION: Varian's HyperSight MAR reconstruction algorithm shows a reduction in metal artifact metrics, motivating the use of MAR reconstruction for patients with metal implants.

2.
Epilepsia ; 64(4): 1061-1073, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36495145

RESUMO

OBJECTIVE: Infantile spasms is an epileptic encephalopathy of childhood, and its pathophysiology is largely unknown. We generated a heterozygous knock-in mouse with the human infantile spasms-associated de novo mutation GABRB3 (c.A328G, p.N110D) to investigate its molecular mechanisms and to establish the Gabrb3+/N110D knock-in mouse as a model of infantile spasms syndrome. METHODS: We used electroencephalography (EEG) and video monitoring to characterize seizure types, and a suite of behavioral tests to identify neurological and behavioral impairment in Gabrb3+/N110D knock-in mice. Miniature inhibitory postsynaptic currents (mIPSCs) were recorded from layer V/VI pyramidal neurons in somatosensory cortex, and extracellular multi-unit recordings from the ventral basal nucleus of the thalamus in a horizontal thalamocortical slice were used to assess spontaneous thalamocortical oscillations. RESULTS: The infantile spasms-associated human de novo mutation GABRB3 (c.A328G, p.N110D) caused epileptic spasms early in development and multiple seizure types in adult Gabrb3+/N110D knock-in mice. Signs of neurological impairment, anxiety, hyperactivity, social impairment, and deficits in spatial learning and memory were also observed. Gabrb3+/N110D mice had reduced cortical mIPSCs and increased duration of spontaneous oscillatory firing in the somatosensory thalamocortical circuit. SIGNIFICANCE: The Gabrb3+/N110D knock-in mouse has epileptic spasms, seizures, and other neurological impairments that are consistent with infantile spasms syndrome in patients. Multiple seizure types and abnormal behaviors indicative of neurological impairment both early and late in development suggest that Gabrb3+/N110D mice can be used to study the pathophysiology of infantile spasms. Reduced cortical inhibition and increased duration of thalamocortical oscillatory firing suggest perturbations in thalamocortical circuits.


Assuntos
Espasmos Infantis , Humanos , Camundongos , Animais , Espasmos Infantis/genética , Receptores de GABA-A/genética , Convulsões , Células Piramidais , Eletroencefalografia , Síndrome , Espasmo
3.
Neurosurg Rev ; 46(1): 270, 2023 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-37843688

RESUMO

Chronic subdural hematoma (CSDH) is a common neurosurgical condition. Surgical evacuation has remained the primary treatment despite many advancements in the endovascular field. Regardless, recurrence requiring reoperation is commonly observed during the postoperative follow-up. Herein, we aimed to investigate risk factors for recurrence after surgical evacuation. A review of MEDLINE, EMBASE, Web of Science, and Scopus was conducted using the designed search string. Studies were reviewed based on the predefined eligibility criteria. Data regarding sixty potential risk factors along with operational information were extracted for analysis. A meta-analysis using the random-effect model was conducted, and each risk factor affecting the postoperative recurrence of CSDH was then evaluated and graded. A total of 198 records met the eligibility criteria. A total number of 8523 patients with recurrent CSDH and 56,096 with non-recurrent CSDH were included in the study. The recurrence rate after surgical evacuation was 12%. Fifteen preoperative, nine radiologic, four hematoma-related, and three operative and postoperative factors were associated with recurrence. Risk factors associated with recurrence after surgical evacuation are important in neurosurgical decision-making and treatment planning. Found risk factors in this study may be used as the basis for pre-operative risk assessment to choose patients who would benefit the most from surgical evacuation.


Assuntos
Hematoma Subdural Crônico , Humanos , Hematoma Subdural Crônico/cirurgia , Hematoma Subdural Crônico/etiologia , Craniotomia , Fatores de Risco , Drenagem/efeitos adversos , Reoperação , Recidiva , Resultado do Tratamento
4.
BJOG ; 129(7): 1133-1139, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35015334

RESUMO

OBJECTIVE: To review the effect of the COVID-19 pandemic on the diagnosis of cervical cancer and model the impact on workload over the next 3 years. DESIGN: A retrospective, control, cohort study. SETTING: Six cancer centres in the North of England representing a combined population of 11.5 million. METHODS: Data were collected retrospectively for all diagnoses of cervical cancer during May-October 2019 (Pre-COVID cohort) and May-October 2020 (COVID cohort). Data were used to generate tools to forecast case numbers for the next 3 years. MAIN OUTCOME MEASURES: Histology, stage, presentation, onset of symptoms, investigation and type of treatment. Patients with recurrent disease were excluded. RESULTS: 406 patients were registered across the study periods; 233 in 2019 and 173 in 2020, representing a 25.7% (n = 60) reduction in absolute numbers of diagnoses. This was accounted for by a reduction in the number of low stage cases (104 in 2019 to 77 in 2020). Adding these data to the additional cases associated with a temporary cessation in screening during the pandemic allowed development of forecasts, suggesting that over the next 3 years there would be 586, 228 and 105 extra cases of local, regional and distant disease, respectively, throughout England. Projection tools suggest that increasing surgical capacity by two or three cases per month per centre would eradicate this excess by 12 months and 7 months, respectively. CONCLUSIONS: There is likely to be a significant increase in cervical cancer cases presenting over the next 3 years. Increased surgical capacity could mitigate this with little increase in morbidity or mortality. TWEETABLE ABSTRACT: Covid will result in 919 extra cases of cervical cancer in England alone. Effects can be mitigated by increasing surgical capacity.


Assuntos
COVID-19 , Neoplasias do Colo do Útero , COVID-19/epidemiologia , Estudos de Coortes , Inglaterra/epidemiologia , Feminino , Humanos , Pandemias , Estudos Retrospectivos , Neoplasias do Colo do Útero/diagnóstico , Neoplasias do Colo do Útero/epidemiologia , Neoplasias do Colo do Útero/patologia
5.
Cereb Cortex ; 31(2): 768-784, 2021 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-32930324

RESUMO

Epileptic activity in genetic generalized epilepsy (GGE) patients preferentially appears during sleep and its mechanism remains unknown. Here, we found that sleep-like slow-wave oscillations (0.5 Hz SWOs) potentiated excitatory and inhibitory synaptic currents in layer V cortical pyramidal neurons from wild-type (wt) mouse brain slices. In contrast, SWOs potentiated excitatory, but not inhibitory, currents in cortical neurons from a heterozygous (het) knock-in (KI) Gabrg2+Q/390X model of Dravet epilepsy syndrome. This created an imbalance between evoked excitatory and inhibitory currents to effectively prompt neuronal action potential firings. Similarly, physiologically similar up-/down-state induction (present during slow-wave sleep) in cortical neurons also potentiated excitatory synaptic currents within brain slices from wt and het KI mice. Moreover, this state-dependent potentiation of excitatory synaptic currents entailed some signaling pathways of homeostatic synaptic plasticity. Consequently, in het KI mice, in vivo SWO induction (using optogenetic methods) triggered generalized epileptic spike-wave discharges (SWDs), being accompanied by sudden immobility, facial myoclonus, and vibrissa twitching. In contrast, in wt littermates, SWO induction did not cause epileptic SWDs and motor behaviors. To our knowledge, this is the first mechanism to explain why epileptic SWDs preferentially happen during non rapid eye-movement sleep and quiet-wakefulness in human GGE patients.


Assuntos
Epilepsia Generalizada/genética , Epilepsia Generalizada/fisiopatologia , Receptores de GABA-A/genética , Convulsões/fisiopatologia , Sinapses , Ácido gama-Aminobutírico , Potenciais de Ação , Animais , Eletroencefalografia , Fenômenos Eletrofisiológicos , Potenciais Pós-Sinápticos Excitadores , Feminino , Masculino , Camundongos , Movimento , Plasticidade Neuronal , Optogenética , Transdução de Sinais , Sono , Sono REM , Sono de Ondas Lentas , Vibrissas
6.
J Appl Clin Med Phys ; 23(11): e13765, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36052983

RESUMO

Class solution template trajectories are used clinically for efficiency, safety, and reproducibility. The aim was to develop class solutions for single cranial metastases radiotherapy/radiosurgery based on intracranial target positioning and compare to patient-specific trajectories in the context of 4π optimization. Template trajectories were constructed based on the open-source Montreal Neurological Institute (MNI) average brain. The MNI brain was populated with evenly spaced spherical target volumes (2 cm diameter, N = 243) and organs-at-risk (OARs) were identified. Template trajectories were generated for six anatomical regions (frontal, medial, and posterior, each with laterality dependence) based on previously published 4π optimization methods. Volumetric modulated arc therapy (VMAT) treatment plans generated using anatomically informed template 4π trajectories and patientspecific 4π trajectories were compared against VMAT plans from a standard four-arc template. Four-arc optimization techniques were compared to the standard VMAT template by placing three spherical targets in each of six anatomical regions of a test patient. This yielded 54 plans to compare various plan quality metrics. Increasing plan technique complexity, the total number of OAR maximum dose reductions compared to the standard arc template for the 6 anatomical classes was 4+/-2 (OFIXEDc) and 7+/-2 (OFIXEDi). In 65.6% of all cases, optimized fixed-couch positions outperformed the standard-arc template. Of the three comparisons, the most complex (OFIXEDi) showed the greatest statistical significance compared to the least complex (VMATi) across 12 plan quality metrics of maximum dose to each OAR, V12Gy, total plan Monitor Units, conformity index, and gradient index (p < 0.00417). In approximately 70% of all cases, 4π optimization methods outperformed the standard-arc template in terms of maximum dose reduction to OAR, by exclusively changing the arc geometry. We conclude that a tradeoff exists between complexity of a class solution methodology compared to patient-specific methods for arc selection, in the context of plan quality improvement.


Assuntos
Radiocirurgia , Radioterapia de Intensidade Modulada , Humanos , Radiocirurgia/métodos , Dosagem Radioterapêutica , Planejamento da Radioterapia Assistida por Computador/métodos , Reprodutibilidade dos Testes , Radioterapia de Intensidade Modulada/métodos
7.
J Appl Clin Med Phys ; 22(12): 72-86, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34679247

RESUMO

PURPOSE: To investigate the possible advantages of using 4pi-optimized arc trajectories in stereotactic body radiation therapy of ventricular tachycardia (VT-SBRT) to minimize exposure of healthy tissues. METHODS AND MATERIALS: Thorax computed tomography (CT) data for 15 patients were used for contouring organs at risk (OARs) and defining realistic planning target volumes (PTVs). A conventional trajectory plan, defined as two full coplanar arcs was compared to an optimized-trajectory plan provided by a 4pi algorithm that penalizes geometric overlap of PTV and OARs in the beam's-eye-view. A single fraction of 25 Gy was prescribed to the PTV in both plans and a comparison of dose sparing to OARs was performed based on comparisons of maximum, mean, and median dose. RESULTS: A significant average reduction in maximum dose was observed for esophagus (18%), spinal cord (26%), and trachea (22%) when using 4pi-optimized trajectories. Mean doses were also found to decrease for esophagus (19%), spinal cord (33%), skin (18%), liver (59%), lungs (19%), trachea (43%), aorta (11%), inferior vena cava (25%), superior vena cava (33%), and pulmonary trunk (26%). A median dose reduction was observed for esophagus (40%), spinal cord (48%), skin (36%), liver (72%), lungs (41%), stomach (45%), trachea (53%), aorta (45%), superior vena cava (38%), pulmonary veins (32%), and pulmonary trunk (39%). No significant difference was observed for maximum dose (p = 0.650) and homogeneity index (p = 0.156) for the PTV. Average values of conformity number were 0.86 ± 0.05 and 0.77 ± 0.09 for the conventional and 4pi optimized plans respectively. CONCLUSIONS: 4pi optimized trajectories provided significant reduction to mean and median doses to cardiac structures close to the target but did not decrease maximum dose. Significant improvement in maximum, mean and median doses for noncardiac OARs makes 4pi optimized trajectories a suitable delivery technique for treating VT.


Assuntos
Radiocirurgia , Radioterapia de Intensidade Modulada , Taquicardia Ventricular , Humanos , Órgãos em Risco , Dosagem Radioterapêutica , Planejamento da Radioterapia Assistida por Computador , Veia Cava Superior
8.
Epilepsia ; 61(10): 2301-2312, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32944937

RESUMO

OBJECTIVE: Neuroinflammation is a major theme in epilepsy, which has been characterized in acquired epilepsy but is poorly understood in genetic epilepsy. γ-Aminobutyric acid type A receptor subunit gene mutations are significant causes of epilepsy, and we have studied the pathophysiology directly resulting from defective receptor channels. Here, we determined the proinflammatory factors in a genetic mouse model, the Gabrg2+/Q390X knockin (KI). We have identified increased cytokines in multiple brain regions of the KI mouse throughout different developmental stages and propose that accumulation of the trafficking-deficient mutant protein may increase neuroinflammation, which would be a novel mechanism for genetic epilepsy. METHODS: We used enzyme-linked immunosorbent assay, immunoprecipitation, nuclei purification, immunoblot, immunohistochemistry, and confocal microscopy to characterize increased neuroinflammation and its potential causes in a Gabrg2+/Q390X KI mouse and a Gabrg2+/- knockout (KO) mouse, each associated with a different epilepsy syndrome with different severities. RESULTS: We found that proinflammatory cytokines such as tumor necrosis factor alpha, interleukin 1-beta (IL-1ß), and IL-6 were increased in the KI mice but not in the KO mice. A major underlying basis for the discrepancy in cytokine expression between the two mouse models is likely chronic mutant protein accumulation and endoplasmic reticulum (ER) stress. The presence of mutant protein dampened cytokine induction upon further cellular stimulation or external stress such as elevated temperature. Pharmacological induction of ER stress upregulated cytokine expression in the wild-type and KO but not in the KI mice. The increased cytokine expression was independent of seizure occurrence, because it was upregulated in both mice and cultured neurons. SIGNIFICANCE: Together, these data demonstrate a novel pathophysiology for genetic epilepsy, increased neuroinflammation, which is a common mechanism for acquired epilepsy. The findings thus provide the first link of neuroinflammation between genetic epilepsy associated with an ion channel gene mutation and acquired epilepsy.


Assuntos
Citocinas/genética , Citocinas/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Epilepsia/genética , Epilepsia/metabolismo , Receptores de GABA-A/genética , Animais , Células Cultivadas , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Modelos Animais de Doenças , Epilepsia/patologia , Feminino , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de GABA-A/deficiência
9.
Brain ; 142(7): 1938-1954, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31056671

RESUMO

We performed next generation sequencing on 1696 patients with epilepsy and intellectual disability using a gene panel with 480 epilepsy-related genes including all GABAA receptor subunit genes (GABRs), and we identified six de novo GABR mutations, two novel GABRA5 mutations (c.880G>T, p.V294F and c.1238C>T, p.S413F), two novel GABRA1 mutations (c.778C>T, p.P260S and c.887T>C, p.L296S/c.944G>T, p.W315L) and two known GABRA1 mutations (c.335G>A, p.R112Q and c.343A>G, p.N115D) in six patients with intractable early onset epileptic encephalopathy. The α5(V294F and S413F) and α1(P260S and L296S/W315L) subunit residue substitutions were all in transmembrane domains, while the α1(R112Q and N115R) subunit residue substitutions were in the N-terminal GABA binding domain. Using multidisciplinary approaches, we compared effects of mutant GABAA receptor α5 and α1 subunits on the properties of recombinant α5ß3γ2 and α1ß3γ2 GABAA receptors in both neuronal and non-neuronal cells and characterized their effects on receptor clustering, biogenesis and channel function. GABAA receptors containing mutant α5 and α1 subunits all had reduced cell surface and total cell expression with altered endoplasmic reticulum processing, impaired synaptic clustering, reduced GABAA receptor function and decreased GABA binding potency. Our study identified GABRA5 as a causative gene for early onset epileptic encephalopathy and expands the mutant GABRA1 phenotypic spectrum, supporting growing evidence that defects in GABAergic neurotransmission contribute to early onset epileptic encephalopathy phenotypes.


Assuntos
Epilepsia/genética , Deficiência Intelectual/genética , Receptores de GABA-A/genética , Sinapses/genética , Criança , Pré-Escolar , Epilepsia/complicações , Feminino , Predisposição Genética para Doença/genética , Humanos , Deficiência Intelectual/complicações , Masculino , Potenciais da Membrana/fisiologia , Potenciais Pós-Sinápticos em Miniatura/fisiologia , Mutação , Cultura Primária de Células , Receptores de GABA-A/biossíntese , Receptores de GABA-A/metabolismo , Receptores de GABA-A/fisiologia , Sinapses/fisiologia , Adulto Jovem , Ácido gama-Aminobutírico/metabolismo
10.
Brain ; 142(10): 3028-3044, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31435640

RESUMO

GABRB3 is highly expressed early in the developing brain, and its encoded ß3 subunit is critical for GABAA receptor assembly and trafficking as well as stem cell differentiation in embryonic brain. To date, over 400 mutations or variants have been identified in GABRB3. Mutations in GABRB3 have been increasingly recognized as a major cause for severe paediatric epilepsy syndromes such as Lennox-Gastaut syndrome, Dravet syndrome and infantile spasms with intellectual disability as well as relatively mild epilepsy syndromes such as childhood absence epilepsy. There is no plausible molecular pathology for disease phenotypic heterogeneity. Here we used a very high-throughput flow cytometry assay to evaluate the impact of multiple human mutations in GABRB3 on receptor trafficking. In this study we found that surface expression of mutant ß3 subunits is variable. However, it was consistent that surface expression of partnering γ2 subunits was lower when co-expressed with mutant than with wild-type subunits. Because γ2 subunits are critical for synaptic GABAA receptor clustering, this provides an important clue for understanding the pathophysiology of GABRB3 mutations. To validate our findings further, we obtained an in-depth comparison of two novel mutations [GABRB3 (N328D) and GABRB3 (E357K)] associated with epilepsy with different severities of epilepsy phenotype. GABRB3 (N328D) is associated with the relatively severe Lennox-Gastaut syndrome, and GABRB3 (E357K) is associated with the relatively mild juvenile absence epilepsy syndrome. With functional characterizations in both heterologous cells and rodent cortical neurons by patch-clamp recordings, confocal microscopy and immunoblotting, we found that both the GABRB3 (N328D) and GABRB3 (E357K) mutations reduced total subunit expression in neurons but not in HEK293T cells. Both mutant subunits, however, were reduced on the cell surface and in synapses, but the Lennox-Gastaut syndrome mutant ß3 (N328D) subunit was more reduced than the juvenile absence epilepsy mutant ß3 (E357K) subunit. Interestingly, both mutant ß3 subunits impaired postsynaptic clustering of wild-type GABAA receptor γ2 subunits and prevented γ2 subunits from incorporating into GABAA receptors at synapses, although by different cellular mechanisms. Importantly, wild-type γ2 subunits were reduced and less clustered at inhibitory synapses in Gabrb3+/- knockout mice. This suggests that impaired receptor localization to synapses is a common pathophysiological mechanism for GABRB3 mutations, although the extent of impairment may be different among mutant subunits. The study thus identifies the novel mechanism of impaired targeting of receptors containing mutant ß3 subunits and provides critical insights into understanding how GABRB3 mutations produce severe epilepsy syndromes and epilepsy phenotypic heterogeneity.


Assuntos
Epilepsia/genética , Receptores de GABA-A/genética , Animais , Encéfalo/embriologia , Linhagem Celular , Membrana Celular/metabolismo , Criança , Pré-Escolar , Análise por Conglomerados , Epilepsia/metabolismo , Síndromes Epilépticas/genética , Feminino , Citometria de Fluxo/métodos , Células HEK293 , Humanos , Masculino , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Knockout , Mutação/genética , Técnicas de Patch-Clamp , Fenótipo , Subunidades Proteicas/genética , Transporte Proteico , Ratos , Receptores de GABA-A/metabolismo
11.
J Neurosci ; 38(11): 2818-2831, 2018 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-29440552

RESUMO

Hyperekplexia, an inherited neuronal disorder characterized by exaggerated startle responses with unexpected sensory stimuli, is caused by dysfunction of glycinergic inhibitory transmission. From analysis of newly identified human hyperekplexia mutations in the glycine receptor (GlyR) α1 subunit, we found that an alanine-to-proline missense mutation (A384P) resulted in substantially higher desensitization level and lower agonist sensitivity of homomeric α1 GlyRs when expressed in HEK cells. The incorporation of the ß subunit fully reversed the reduction in agonist sensitivity and partially reversed the desensitization of α1A384P The heteromeric α1A384Pß GlyRs showed enhanced desensitization but unchanged agonist-induced maximum responses, surface expression, main channel conductance, and voltage dependence compared with that of the wild-type α1ß (α1WTß) GlyRs. Coexpression of the R392H and A384P mutant α1 subunits, which mimic the expression of the compound heterozygous mutation in a hyperekplexia patient, resulted in channel properties similar to those with α1A384P subunit expression alone. In comparison, another human hyperekplexia mutation α1P250T, which was previously reported to enhance desensitization, caused a strong reduction in maximum currents in addition to the altered desensitization. These results were further confirmed by overexpression of α1P250T or α1A384P subunits in cultured neurons isolated from SD rats of either sex. Moreover, the IPSC-like responses of cells expressing α1A384Pß induced by repeated glycine pulses showed a stronger frequency-dependent reduction than those expressing α1WTß. Together, our findings demonstrate that A384 is associated with the desensitization site of the α1 subunit and its proline mutation produced enhanced desensitization of GlyRs, which contributes to the pathogenesis of human hyperekplexia.SIGNIFICANCE STATEMENT Human startle disease is caused by impaired synaptic inhibition in the brainstem and spinal cord, which is due to either direct loss of GlyR channel function or reduced number of synaptic GlyRs. Considering that fast decay kinetics of GlyR-mediated inhibitory synaptic responses, the question was raised whether altered desensitization of GlyRs will cause dysfunction of glycine transmission and disease phenotypes. Here, we found that the α1 subunit mutation A384P, identified from startle disease patients, results in enhanced desensitization and leads to rapidly decreasing responses in the mutant GlyRs when they are activated repeatedly by the synaptic-like simulation. These observations suggest that the enhanced desensitization of postsynaptic GlyRs could be the primary pathogenic mechanism of human startle disease.


Assuntos
Rigidez Muscular/genética , Mutação de Sentido Incorreto/genética , Receptores de Glicina/genética , Animais , Biotinilação , Células Cultivadas , Potenciais Pós-Sinápticos Excitadores/genética , Feminino , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Glicina/farmacologia , Células HEK293 , Humanos , Masculino , Técnicas de Patch-Clamp , Prolina/genética , Ratos , Ratos Sprague-Dawley
12.
Epilepsia ; 60(6): 1137-1149, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31087664

RESUMO

OBJECTIVE: γ-Aminobutyric acid type A (GABAA ) receptor subunit gene mutations are significant causes of epilepsy, which are often accompanied by various neuropsychiatric comorbidities, but the underlying mechanisms are unclear. It has been suggested that the comorbidities are caused by seizures, as the comorbidities often present in severe epilepsy syndromes. However, findings from both humans and animal models argue against this conclusion. Mutations in the GABAA receptor γ2 subunit gene GABRG2 have been associated with anxiety alone or with severe epilepsy syndromes and comorbid anxiety, suggesting that a core molecular defect gives rise to the phenotypic spectrum. Here, we determined the pathophysiology of comorbid anxiety in GABRG2 loss-of-function epilepsy syndromes, identified the central nucleus of the amygdala (CeA) as a primary site for epilepsy comorbid anxiety, and demonstrated a potential rescue of comorbid anxiety via neuromodulation of CeA neurons. METHODS: We used brain slice recordings, subcellular fractionation with Western blot, immunohistochemistry, confocal microscopy, and a battery of behavior tests in combination with a chemogenetic approach to characterize anxiety and its underlying mechanisms in a Gabrg2+/Q390X knockin mouse and a Gabrg2+/- knockout mouse, each associated with a different epilepsy syndrome. RESULTS: We found that impaired GABAergic neurotransmission in CeA underlies anxiety in epilepsy, which is due to reduced GABAA receptor subunit expression resulting from the mutations. Impaired GABAA receptor expression reduced GABAergic neurotransmission in CeA, but not in basolateral amygdala. Activation or inactivation of inhibitory neurons using a chemogenetic approach in CeA alone modulated anxietylike behaviors. Similarly, pharmacological enhancement of GABAergic signaling via γ2 subunit-containing receptors relieved the anxiety. SIGNIFICANCE: Together, these data demonstrate the molecular basis for a comorbidity of epilepsy, anxiety, and suggest that impaired GABAA receptor function in CeA due to a loss-of-function mutation could at least contribute to anxiety. Modulation of CeA neurons could cause or suppress anxiety, suggesting a potential use of CeA neurons as therapeutic targets for treatment of anxiety in addition to traditional pharmacological approaches.


Assuntos
Tonsila do Cerebelo/fisiopatologia , Ansiedade/complicações , Ansiedade/genética , Epilepsia/complicações , Epilepsia/genética , Receptores de GABA-A/genética , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Ansiolíticos/farmacologia , Ansiedade/tratamento farmacológico , Comportamento Animal , Comorbidade , Eletroencefalografia , Epilepsia/fisiopatologia , Potenciais Pós-Sinápticos Excitadores , Humanos , Camundongos , Camundongos Knockout , Neurônios/efeitos dos fármacos , Receptores de GABA-A/deficiência , Transmissão Sináptica , Ácido gama-Aminobutírico/fisiologia
13.
J Physiol ; 596(18): 4475-4495, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30019335

RESUMO

KEY POINTS: Physiologically relevant combinations of recombinant GABAA receptor (GABAR) subunits were expressed in HEK293 cells. Using whole-cell voltage clamp and rapid drug application, we measured the GABAR-subtype-specific properties to convey either synaptic or extrasynaptic signalling in a range of physiological contexts. α4ßδ GABARs are optimally tuned to submicromolar tonic GABA and transient surges of micromolar GABA concentrations. α5ß2γ2l GABARs are better suited to higher tonic GABA levels, but also convey robust responses to brief synaptic and perisynaptic GABA fluctuations. α1ß2/3δ GABARs function well at prolonged, micromolar (>2 µm) GABA levels, but not to low tonic (<1 µm GABA) or synaptic/transient GABAergic signalling. These results help illuminate the context- and isoform-specific modes of GABAergic signalling in the brain. ABSTRACT: GABAA receptors (GABARs) mediate a remarkable diversity of signalling modalities in vivo. Yet most published work characterizing responses to GABA has focused on the properties needed to convey fast, phasic synaptic inhibition. We therefore aimed to characterize the most prevalent (α4ßδ, α5ß3γ2L) and least prevalent (α1ß2δ) non-synaptic GABAR currents, using whole-cell voltage clamp recordings of recombinant GABAR expressed in HEK293 cells and drug application protocols to recapitulate the GABA concentration profiles occurring during both fast synaptic and slow extrasynaptic signalling. We found that α4ßδ GABARs were very sensitive to submicromolar GABA, with a rank order potency of α4ß2δ ≥ α4ß1δ ≈ α4ß3δ GABARs. In comparison, the GABA EC50 was up to 20 times higher for α1ß2γ2L GABARs, with α1ß2δ and α5ß3γ2L GABARs having intermediate GABA potency. Both α4ßδ and α5ß3γ2L GABAR currents exhibited slow, but substantial, desensitization as well as prolonged rates of deactivation. These GABAR current properties defined distinct 'dynamic ranges' of responsiveness to changing GABA for α4ß2δ (0.1-1 µm), α5ß3γ2L (0.5-7 µm) and α1ß2γ2L (0.6-9 µm) GABARs. Finally, α1ß2δ GABARs were notable for their relative lack of desensitization and extremely quick deactivation. In summary, our results help delineate the roles that specific GABARs may play in mediating non-synaptic GABA signals. Since ambient GABA levels may be altered during development as well as by drugs and disease states, these findings may help future efforts to understand disrupted inhibition underlying a variety of neurological illnesses, such as epilepsy.


Assuntos
Receptores de GABA-A/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , GABAérgicos/farmacologia , Células HEK293 , Humanos , Isoformas de Proteínas/metabolismo , Ratos , Receptores de GABA-A/química , Ácido gama-Aminobutírico/farmacologia
14.
Hum Mol Genet ; 25(15): 3192-3207, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27340224

RESUMO

Genetic epilepsy is a common disorder with phenotypic variation, but the basis for the variation is unknown. Comparing the molecular pathophysiology of mutations in the same epilepsy gene may provide mechanistic insights into the phenotypic heterogeneity. GABRG2 is an established epilepsy gene, and mutations in it produce epilepsy syndromes with varying severities. The disease phenotype in some cases may be caused by simple loss of subunit function (functional haploinsufficiency), while others may be caused by loss-of-function plus dominant negative suppression and other cellular toxicity. Detailed molecular defects and the corresponding seizures and related comorbidities resulting from haploinsufficiency and dominant negative mutations, however, have not been compared. Here we compared two mouse models of GABRG2 loss-of-function mutations associated with epilepsy with different severities, Gabrg2+/Q390X knockin (KI) and Gabrg2+/- knockout (KO) mice. Heterozygous Gabrg2+/Q390X KI mice are associated with a severe epileptic encephalopathy due to a dominant negative effect of the mutation, while heterozygous Gabrg2+/- KO mice are associated with mild absence epilepsy due to simple haploinsufficiency. Unchanged at the transcriptional level, KI mice with severe epilepsy had neuronal accumulation of mutant γ2 subunits, reduced remaining functional wild-type subunits in dendrites and synapses, while KO mice with mild epilepsy had no intracellular accumulation of the mutant subunits and unaffected biogenesis of the remaining wild-type subunits. Consequently, KI mice with dominant negative mutations had much less wild-type receptor expression, more severe seizures and behavioural comorbidities than KO mice. This work provides insights into the pathophysiology of epilepsy syndrome heterogeneity and designing mechanism-based therapies.


Assuntos
Comportamento Animal , Epilepsia/genética , Genes Dominantes , Haploinsuficiência , Mutação de Sentido Incorreto , Receptores de GABA-A/genética , Substituição de Aminoácidos , Animais , Modelos Animais de Doenças , Epilepsia/metabolismo , Epilepsia/fisiopatologia , Humanos , Camundongos , Camundongos Knockout , Receptores de GABA-A/metabolismo
15.
Brain ; 140(1): 49-67, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27864268

RESUMO

Epileptic encephalopathies are a devastating group of severe childhood onset epilepsies with medication-resistant seizures and poor developmental outcomes. Many epileptic encephalopathies have a genetic aetiology and are often associated with de novo mutations in genes mediating synaptic transmission, including GABAA receptor subunit genes. Recently, we performed next generation sequencing on patients with a spectrum of epileptic encephalopathy phenotypes, and we identified five novel (A106T, I107T, P282S, R323W and F343L) and one known (R323Q) de novo GABRG2 pathogenic variants (mutations) in eight patients. To gain insight into the molecular basis for how these mutations contribute to epileptic encephalopathies, we compared the effects of the mutations on the properties of recombinant α1ß2γ2L GABAA receptors transiently expressed in HEK293T cells. Using a combination of patch clamp recording, immunoblotting, confocal imaging and structural modelling, we characterized the effects of these GABRG2 mutations on GABAA receptor biogenesis and channel function. Compared with wild-type α1ß2γ2L receptors, GABAA receptors containing a mutant γ2 subunit had reduced cell surface expression with altered subunit stoichiometry or decreased GABA-evoked whole-cell current amplitudes, but with different levels of reduction. While a causal role of these mutations cannot be established directly from these results, the functional analysis together with the genetic information suggests that these GABRG2 variants may be major contributors to the epileptic encephalopathy phenotypes. Our study further expands the GABRG2 phenotypic spectrum and supports growing evidence that defects in GABAergic neurotransmission participate in the pathogenesis of genetic epilepsies including epileptic encephalopathies.


Assuntos
Epilepsia Resistente a Medicamentos/genética , Epilepsia Resistente a Medicamentos/fisiopatologia , Epilepsia/genética , Epilepsia/fisiopatologia , Receptores de GABA-A/genética , Criança , Pré-Escolar , Fenômenos Eletrofisiológicos , Exoma , Feminino , Células HEK293 , Humanos , Masculino , Mutação , Técnicas de Patch-Clamp , Fenótipo
16.
J Med Genet ; 54(3): 202-211, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27789573

RESUMO

BACKGROUND: Early myoclonic encephalopathy (EME), a disease with a devastating prognosis, is characterised by neonatal onset of seizures and massive myoclonus accompanied by a continuous suppression-burst EEG pattern. Three genes are associated with EMEs that have metabolic features. Here, we report a pathogenic mutation of an ion channel as a cause of EME for the first time. METHODS: Sequencing was performed for 214 patients with epileptic seizures using a gene panel with 109 genes that are known or suspected to cause epileptic seizures. Functional assessments were demonstrated by using electrophysiological experiments and immunostaining for mutant γ-aminobutyric acid-A (GABAA) receptor subunits in HEK293T cells. RESULTS: We discovered a de novo heterozygous missense mutation (c.859A>C [p.Thr287Pro]) in the GABRB2-encoded ß2 subunit of the GABAA receptor in an infant with EME. No GABRB2 mutations were found in three other EME cases or in 166 patients with infantile spasms. GABAA receptors bearing the mutant ß2 subunit were poorly trafficked to the cell membrane and prevented γ2 subunits from trafficking to the cell surface. The peak amplitudes of currents from GABAA receptors containing only mutant ß2 subunits were smaller than that of those from receptors containing only wild-type ß2 subunits. The decrease in peak current amplitude (96.4% reduction) associated with the mutant GABAA receptor was greater than expected, based on the degree to which cell surface expression was reduced (66% reduction). CONCLUSION: This mutation has complex functional effects on GABAA receptors, including reduction of cell surface expression and attenuation of channel function, which would significantly perturb GABAergic inhibition in the brain.


Assuntos
Síndrome de Opsoclonia-Mioclonia/genética , Receptores de GABA-A/genética , Convulsões/genética , Espasmos Infantis/genética , Encéfalo/diagnóstico por imagem , Encéfalo/fisiopatologia , Cristalografia por Raios X , Eletroencefalografia , Células HEK293 , Humanos , Lactente , Masculino , Modelos Moleculares , Mutação de Sentido Incorreto , Síndrome de Opsoclonia-Mioclonia/fisiopatologia , Receptores de GABA-A/química , Convulsões/fisiopatologia , Espasmos Infantis/fisiopatologia
17.
BMC Public Health ; 18(1): 859, 2018 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-29996792

RESUMO

BACKGROUND: South Africa remains the global epicentre of HIV infection, and adolescent women have the highest incidence of HIV in the country. South Africa also has high rates of alcohol and other drug (AOD) use, violence, and gender inequality. Violence converges with AOD use, gender inequities and other disparities, such as poverty, to increase sexual risk and poor educational attainment for adolescent women. This study seeks to test the efficacy of peer recruitment and cofacilitation of the Young Women's Health CoOp (YWHC), a comprehensive gender-focused intervention to reduce HIV risk behaviours and increase the uptake of HIV counselling and testing (HCT) among out-of-school, adolescent women who use AODs. The YWHC is facilitated by local research staff and supported by peers. METHODS: This cluster-randomised trial is enrolling participants into two arms: a control arm that receives standard HCT, and an intervention arm that receives the YWHC. Participants are enrolled from 24 economically disadvantaged communities in Cape Town, South Africa. These geographically distinct communities serve as clusters that are the units of randomisation. This study uses adolescent peer role models and research field staff to recruit marginalised adolescent women. At baseline, participants complete a questionnaire and biological testing for HIV, recent AOD use, and pregnancy. The core intervention is delivered in the month following enrollment, with linkages to health services and educational programmes available to participants throughout the follow-up period. Follow-up interviews and biological testing are conducted at 6 and 12 months post enrollment. DISCUSSION: The study findings will increase knowledge of the efficacy of a comprehensive HCT, gender-focused programme in reducing AOD use, victimisation, and sexual risk behaviour and increase uptake services for out-of-school, adolescent women who use AODs. The trial results could lead to wider implementation of the YWHC for vulnerable adolescent women, a key population often neglected in health services. TRIAL REGISTRATION: Trial registration no: NCT02974998 , November 29, 2016.


Assuntos
Aconselhamento , Infecções por HIV/diagnóstico , Infecções por HIV/terapia , Assunção de Riscos , Populações Vulneráveis/estatística & dados numéricos , Adolescente , Adulto , Análise por Conglomerados , Escolaridade , Feminino , Infecções por HIV/prevenção & controle , Humanos , Grupo Associado , Pobreza , Projetos de Pesquisa , África do Sul , Adulto Jovem
18.
J Biol Chem ; 291(39): 20440-61, 2016 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-27493204

RESUMO

The subunit stoichiometry and arrangement of synaptic αßγ GABAA receptors are generally accepted as 2α:2ß:1γ with a ß-α-γ-ß-α counterclockwise configuration, respectively. Whether extrasynaptic αßδ receptors adopt the analogous ß-α-δ-ß-α subunit configuration remains controversial. Using flow cytometry, we evaluated expression levels of human recombinant γ2 and δ subunits when co-transfected with α1 and/or ß2 subunits in HEK293T cells. Nearly identical patterns of γ2 and δ subunit expression were observed as follows: both required co-transfection with α1 and ß2 subunits for maximal expression; both were incorporated into receptors primarily at the expense of ß2 subunits; and both yielded similar FRET profiles when probed for subunit adjacency, suggesting similar underlying subunit arrangements. However, because of a slower rate of δ subunit degradation, 10-fold less δ subunit cDNA was required to recapitulate γ2 subunit expression patterns and to eliminate the functional signature of α1ß2 receptors. Interestingly, titrating γ2 or δ subunit cDNA levels progressively altered GABA-evoked currents, revealing more than one kinetic profile for both αßγ and αßδ receptors. This raised the possibility of alternative receptor isoforms, a hypothesis confirmed using concatameric constructs for αßγ receptors. Taken together, our results suggest a limited cohort of alternative subunit arrangements in addition to canonical ß-α-γ/δ-ß-α receptors, including ß-α-γ/δ-α-α receptors at lower levels of γ2/δ expression and ß-α-γ/δ-α-γ/δ receptors at higher levels of expression. These findings provide important insight into the role of GABAA receptor subunit under- or overexpression in disease states such as genetic epilepsies.


Assuntos
Regulação da Expressão Gênica/fisiologia , Potenciais da Membrana/fisiologia , Subunidades Proteicas/biossíntese , Receptores de GABA/biossíntese , Epilepsia/genética , Epilepsia/metabolismo , Epilepsia/fisiopatologia , Citometria de Fluxo , Células HEK293 , Humanos , Subunidades Proteicas/genética , Receptores de GABA/genética
19.
Ann Neurol ; 79(5): 806-825, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26950270

RESUMO

OBJECTIVE: The Epi4K Consortium recently identified 4 de novo mutations in the γ-aminobutyric acid type A (GABAA ) receptor ß3 subunit gene GABRB3 and 1 in the ß1 subunit gene GABRB1 in children with one of the epileptic encephalopathies (EEs) Lennox-Gastaut syndrome (LGS) and infantile spasms (IS). Because the etiology of EEs is often unknown, we determined the impact of GABRB mutations on GABAA receptor function and biogenesis. METHODS: GABAA receptor α1 and γ2L subunits were coexpressed with wild-type and/or mutant ß3 or ß1 subunits in HEK 293T cells. Currents were measured using whole cell and single channel patch clamp techniques. Surface and total expression levels were measured using flow cytometry. Potential structural perturbations in mutant GABAA receptors were explored using structural modeling. RESULTS: LGS-associated GABRB3(D120N, E180G, Y302C) mutations located at ß+ subunit interfaces reduced whole cell currents by decreasing single channel open probability without loss of surface receptors. In contrast, IS-associated GABRB3(N110D) and GABRB1(F246S) mutations at ß- subunit interfaces produced minor changes in whole cell current peak amplitude but altered current deactivation by decreasing or increasing single channel burst duration, respectively. GABRB3(E180G) and GABRB1(F246S) mutations also produced spontaneous channel openings. INTERPRETATION: All 5 de novo GABRB mutations impaired GABAA receptor function by rearranging conserved structural domains, supporting their role in EEs. The primary effect of LGS-associated mutations was reduced GABA-evoked peak current amplitudes, whereas the major impact of IS-associated mutations was on current kinetic properties. Despite lack of association with epilepsy syndromes, our results suggest GABRB1 as a candidate human epilepsy gene. Ann Neurol 2016;79:806-825.

20.
Epilepsia ; 58(8): 1451-1461, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28586508

RESUMO

OBJECTIVE: The mutant γ-aminobutyric acid type A (GABAA ) receptor γ2(Q390X) subunit (Q351X in the mature peptide) has been associated with the epileptic encephalopathy, Dravet syndrome, and the epilepsy syndrome genetic epilepsy with febrile seizures plus (GEFS+). The mutation generates a premature stop codon that results in translation of a stable truncated and misfolded γ2 subunit that accumulates in neurons, forms intracellular aggregates, disrupts incorporation of γ2 subunits into GABAA receptors, and affects trafficking of partnering α and ß subunits. Heterozygous Gabrg2+/Q390X knock-in (KI) mice had reduced cortical inhibition, spike wave discharges on electroencephalography (EEG), a lower seizure threshold to the convulsant drug pentylenetetrazol (PTZ), and spontaneous generalized tonic-clonic seizures. In this proof-of-principal study, we attempted to rescue these deficits in KI mice using a γ2 subunit gene (GABRG2) replacement therapy. METHODS: We introduced the GABRG2 allele by crossing Gabrg2+/Q390X KI mice with bacterial artificial chromosome (BAC) transgenic mice overexpressing HA (hemagglutinin)-tagged human γ2HA subunits, and compared GABAA receptor subunit expression by Western blot and immunohistochemical staining, seizure threshold by monitoring mouse behavior after PTZ-injection, and thalamocortical inhibition and network oscillation by slice recording. RESULTS: Compared to KI mice, adult mice carrying both mutant allele and transgene had increased wild-type γ2 and partnering α1 and ß2/3 subunits, increased miniature inhibitory postsynaptic current (mIPSC) amplitudes recorded from layer VI cortical neurons, reduced thalamocortical network oscillations, and higher PTZ seizure threshold. SIGNIFICANCE: Based on these results we suggest that seizures in a genetic epilepsy syndrome caused by epilepsy mutant γ2(Q390X) subunits with dominant negative effects could be rescued potentially by overexpression of wild-type γ2 subunits.


Assuntos
Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/terapia , Mutação/genética , Subunidades Proteicas/metabolismo , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Animais , Convulsivantes/toxicidade , Estimulação Elétrica , Humanos , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Técnicas de Patch-Clamp , Pentilenotetrazol/toxicidade , Subunidades Proteicas/genética , Células Piramidais/efeitos dos fármacos , Células Piramidais/fisiologia , Córtex Somatossensorial/citologia , Tálamo/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA