Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Mol Neurobiol ; 57(1): 1-10, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31493242

RESUMO

The cation-chloride cotransporters Na+-K+-2Cl--1 (NKCC1) and K+-2Cl--2 (KCC2) critically regulate neuronal responses to gamma-aminobutyric acid (GABA). NKCC1 renders GABA excitatory in immature neurons while expression of KCC2 signals GABA maturation to its inhibitory role. Imbalances in NKCC1/KCC2 alter GABA neurotransmission, which may contribute to hyperexcitability and blunted inhibition in neurocircuitry after neonatal exposure to anesthesia. Thus, we hypothesized that anesthetics may dysregulate NKCC1 and/or KCC2 in developing brain. We exposed postnatal day (PND) 7 mice to sevoflurane or carrier gases and assessed NKCC1 and KCC2 expression across three brain regions 6 h and 24 h after initial exposure. To test differences in behavior, we challenged pups receiving sevoflurane or carrier gases on PND7 with propofol on PND8 and recorded parameters of anesthesia induction and maintenance. Sevoflurane exposure increased cortical NKCC1 at 6 h (p = 0.03) and decreased cortical and hippocampal KCC2 at 24 h (p = 0.009 and p = 0.007, respectively). NKCC1/KCC2 ratio was significantly increased at both 6 h (p = 0.02) and 24 h (p = 0.03) in cortex and at 24 h (p = 0.02) in hippocampus. After propofol challenge on PND8, pups previously exposed to sevoflurane on PND7 regained righting reflex significantly faster than their non-exposed cohort (p < 0.001). Disturbing NKCC1/KCC2 balance may underlie circuit hyperexcitability and contribute to neurodevelopmental impairments we have observed in previous studies of neonatal anesthesia exposure. Human infants previously exposed to anesthesia may require higher concentrations of anesthetic drugs, potentially compounding their susceptibility for neurodevelopmental sequalae.


Assuntos
Encéfalo/metabolismo , Cátions/metabolismo , Cloretos/metabolismo , Membro 2 da Família 12 de Carreador de Soluto/metabolismo , Simportadores/metabolismo , Animais , Animais Recém-Nascidos , Hipocampo/metabolismo , Camundongos , Neurônios/metabolismo , Reflexo de Endireitamento , Sevoflurano/farmacologia , Cotransportadores de K e Cl-
2.
Neurochem Int ; 127: 80-86, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30471325

RESUMO

Each year there are approximately 7000 out of hospital cardiac arrests in the pediatric population, with 30% resuscitation rate and a 6-10% rate of survival to hospital discharge. Survivors of cardiac arrest exhibit learning and memory deficits that are devastating during the school years. Delayed neuronal cell death occurs in the hippocampus following cardiac arrest and likely contributes to memory impairments. Circulating endogenous estrogen in young adult females has been shown to provide protection against ischemic cell death, as does chronic exogenous administration of 17ß-estradiol (E2). Chronic estrogen benefit can have undesirable feminizing effects, particularly in pre-adolescents. Here, we tested if a single-dose of E2 is neuroprotective in our pediatric cardiac arrest mouse model performed in juvenile mice. We subjected P21P25 C57Blk6 male and female mice to 8 min of cardiac arrest followed by cardiopulmonary resuscitation (CA/CPR). This developmental stage preceded the hormonal onset and serum estradiol and testosterone levels were not different in males and females. A single dose of E2 (100µg/kg) or vehicle was administered 30 min after resuscitation. Neuronal cell death measured 3 days after CA/CPR showed reduced hippocampal cell death in E2-treated females, but not males. Benefit of E2 in females was blocked by the P38 MAPK inhibitor, SB203580. Hippocampal-dependent memory function was equally impaired in E2-and vehicle-treated females measured in the contextual fear conditioning task at 7 days. Our findings demonstrate female-specific transient neuroprotection with E2 that does not provide sustained functional benefit.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Estradiol/farmacologia , Parada Cardíaca/tratamento farmacológico , Neuroproteção/efeitos dos fármacos , Animais , Isquemia Encefálica/metabolismo , Modelos Animais de Doenças , Feminino , Masculino , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Fatores Sexuais
3.
Neuroscience ; 284: 555-565, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-25450957

RESUMO

Motor deficits are present in cardiac arrest survivors and injury to cerebellar Purkinje cells (PCs) likely contribute to impairments in motor coordination and post-hypoxic myoclonus. N-Methyl-D-aspartic acid (NMDA) receptor-mediated excitotoxicity is a well-established mechanism of cell death in several brain regions, but the role of NMDA receptors in PC injury remains understudied. Emerging data in cortical and hippocampal neurons indicate that the GluN2A-containing NMDA receptors signal to improve cell survival and GluN2B-containing receptors contribute to neuronal injury. This study compared neuronal injury in the hippocampal CA1 region to that in PCs and investigated the role of NMDA receptors in PC injury in our mouse model of cardiac arrest and cardiopulmonary resuscitation (CA/CPR). Analysis of cell density demonstrated a 24% loss of PCs within 24 h after 8 min CA/CPR and injury stabilized to 33% by 7 days. The subunit promiscuous NMDA receptor antagonist MK-801 protected both CA1 neurons and PCs from ischemic injury following CA/CPR, demonstrating a role for NMDA receptor activation in injury to both brain regions. In contrast, the GluN2B antagonist, Co 101244, had no effect on PC loss while protecting against injury in the CA1 region. These data indicate that ischemic injury to cerebellar PCs progresses via different cell death mechanisms compared to hippocampal CA1 neurons.


Assuntos
Isquemia Encefálica/fisiopatologia , Região CA1 Hipocampal/fisiopatologia , Neurônios/fisiologia , Células de Purkinje/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Isquemia Encefálica/patologia , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/patologia , Calbindinas/metabolismo , Reanimação Cardiopulmonar , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Modelos Animais de Doenças , Maleato de Dizocilpina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Parada Cardíaca , Masculino , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Piperidinas/farmacologia , Células de Purkinje/efeitos dos fármacos , Células de Purkinje/patologia , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Técnicas de Cultura de Tecidos
4.
Eur J Neurosci ; 40(8): 3179-88, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25080203

RESUMO

Global cerebral ischemia following cardiac arrest and cardiopulmonary resuscitation (CA/CPR) causes injury to hippocampal CA1 pyramidal neurons and impairs cognition. Small conductance Ca(2+)-activated potassium channels type 2 (SK2), expressed in CA1 pyramidal neurons, have been implicated as potential protective targets. Here we showed that, in mice, hippocampal long-term potentiation (LTP) was impaired as early as 3 h after recovery from CA/CPR and LTP remained impaired for at least 30 days. Treatment with the SK2 channel agonist 1-Ethyl-2-benzimidazolinone (1-EBIO) at 30 min after CA provided sustained protection from plasticity deficits, with LTP being maintained at control levels at 30 days after recovery from CA/CPR. Minimal changes in glutamate release probability were observed at delayed times after CA/CPR, implicating post-synaptic mechanisms. Real-time quantitative reverse transcriptase-polymerase chain reaction indicated that CA/CPR did not cause a loss of N-methyl-D-aspartate (NMDA) receptor mRNA at 7 or 30 days after CA/CPR. Similarly, no change in synaptic NMDA receptor protein levels was observed at 7 or 30 days after CA/CPR. Further, patch-clamp experiments demonstrated no change in functional synaptic NMDA receptors at 7 or 30 days after CA/CPR. Electrophysiology recordings showed that synaptic SK channel activity was reduced for the duration of experiments performed (up to 30 days) and that, surprisingly, treatment with 1-EBIO did not prevent the CA/CPR-induced loss of synaptic SK channel function. We concluded that CA/CPR caused alterations in post-synaptic signaling that were prevented by treatment with the SK2 agonist 1-EBIO, indicating that activators of SK2 channels may be useful therapeutic agents to prevent ischemic injury and cognitive impairments.


Assuntos
Isquemia Encefálica/fisiopatologia , Hipocampo/fisiopatologia , Potenciação de Longa Duração , Canais de Potássio Ativados por Cálcio de Condutância Baixa/fisiologia , Animais , Benzimidazóis/farmacologia , Isquemia Encefálica/prevenção & controle , Agonistas dos Canais de Cálcio/farmacologia , Hipocampo/efeitos dos fármacos , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de AMPA/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia
6.
J Neurosci Methods ; 222: 34-41, 2014 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-24192226

RESUMO

BACKGROUND: Pediatric sudden cardiac arrest (CA) is an unfortunate and devastating condition, often leading to poor neurologic outcomes. However, little experimental data on the pathophysiology of pediatric CA is currently available due to the scarcity of animal models. NEW METHOD: We developed a novel experimental model of pediatric cardiac arrest and cardiopulmonary resuscitation (CA/CPR) using postnatal day 20-25 mice. Adult (8-12 weeks) and pediatric (P20-25) mice were subjected to 6min CA/CPR. Hippocampal CA1 and striatal neuronal injury were quantified 3 days after resuscitation by hematoxylin and eosin (H&E) and Fluoro-Jade B staining, respectively. RESULTS: Pediatric mice exhibited less neuronal injury in both CA1 hippocampal and striatal neurons compared to adult mice. Increasing ischemia time to 8 min CA/CPR resulted in an increase in hippocampal injury in pediatric mice, resulting in similar damage in adult and pediatric brains. In contrast, striatal injury in the pediatric brain following 6 or 8 min CA/CPR remained extremely low. As observed in adult mice, cardiac arrest causes delayed neuronal death in pediatric mice, with hippocampal CA1 neuronal damage maturing at 72 h after insult. Finally, mild therapeutic hypothermia reduced hippocampal CA1 neuronal injury after pediatric CA/CPR. COMPARISON WITH EXISTING METHOD: This is the first report of a cardiac arrest and CPR model of global cerebral ischemia in mice. CONCLUSIONS: Therefore, the mouse pediatric CA/CPR model we developed is unique and will provide an important new tool to the research community for the study of pediatric brain injury.


Assuntos
Isquemia Encefálica/etiologia , Isquemia Encefálica/patologia , Reanimação Cardiopulmonar , Modelos Animais de Doenças , Parada Cardíaca/complicações , Parada Cardíaca/terapia , Envelhecimento , Animais , Isquemia Encefálica/terapia , Região CA1 Hipocampal/crescimento & desenvolvimento , Região CA1 Hipocampal/patologia , Morte Celular , Corpo Estriado/crescimento & desenvolvimento , Corpo Estriado/patologia , Amarelo de Eosina-(YS) , Fluoresceínas , Parada Cardíaca/patologia , Hematoxilina , Hipotermia Induzida , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/patologia , Pediatria , Fatores de Tempo
7.
Ann Rheum Dis ; 73(1): 56-61, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24067785

RESUMO

OBJECTIVE: The primary objective of the study was to explore safety and tolerability of hyperimmune caprine serum (AIMSPRO) in established diffuse cutaneous systemic sclerosis (SSc). Secondary objectives included assessment of potential efficacy and biological activity and exploration of candidate biomarkers. METHODS: This was a double-blind parallel group randomised placebo-controlled clinical trial. After informed consent 20 patients with established diffuse cutaneous SSc of greater than 3 years duration not receiving immunosuppressive therapy were randomised to receive either active (n=10) or placebo formulation (n=10) by subcutaneous twice weekly injection over 26 weeks. Clinical assessments were evaluated over 26 weeks. RESULTS: There were no safety concerns during this study. Frequency of adverse events was not different between active and placebo groups. Mean modified Rodnan Skin Score (mRSS) fell by 1.4±4.7 units with active treatment but increased by 2.1±6.4 units on placebo when baseline values were compared with 26 weeks and responder analysis showed clinically meaningful improvement in mRSS at 26 weeks in 5 (50%) of actively treated patients compared with 1 (10%) in the control group (p=0.062). PIIINP (µg/L) showed a comparatively larger increase in the treatment group compared with the placebo group, (p=0.0118). CONCLUSIONS: These results confirm tolerability and safety of this novel biological agent in established diffuse SSc. The value of a placebo treated control group in small clinical trials evaluating skin disease in SSc is confirmed. Potential improvement in mRSS and changes in PIIINP in cases receiving active therapy suggest that this intervention may be of clinical benefit and warrants further evaluation.


Assuntos
Soros Imunes/administração & dosagem , Fatores Imunológicos/administração & dosagem , Esclerodermia Difusa/tratamento farmacológico , Esclerodermia Difusa/imunologia , Adulto , Idoso , Animais , Biomarcadores/sangue , Estudos de Viabilidade , Feminino , Cabras , Humanos , Soros Imunes/efeitos adversos , Fatores Imunológicos/efeitos adversos , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Esclerodermia Difusa/patologia , Resultado do Tratamento
8.
QJM ; 106(9): 839-48, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23696678

RESUMO

BACKGROUND: We have studied endothelin-1 (ET-1) levels and ET-1 ligand and receptor tissue expression in scleroderma renal crisis (SRC) and undertaken a pilot open label safety study of bosentan, a non-selective ET-1 receptor antagonist, in SRC [Bosentan in Renal Disease-1 (BIRD-1)]. METHODS: Serum levels of ET-1 were measured in healthy controls (n = 20) or systemic sclerosis (SSc) (n = 80) with or without SRC, including cases of pulmonary arterial hypertension (PAH). Renal biopsies (n = 27) from patients with SRC were stained for endothelin ligand and receptors. Six cases of SRC received 6 months bosentan. Outcome measures were compared with SRC cases managed at our centre from 2000 to 2004 (n = 49). RESULTS: Serum ET-1 was elevated in SRC but less than in PAH. ET-1 and both endothelin A and endothelin B receptor expression was increased in SRC biopsies in glomeruli, interstitium and hallmark vascular lesions of SRC. In the BIRD-1 cohort, serum ET-1 was elevated in all cases at SRC (median healthy controls 0.50 pg/ml; SRC 1.48 pg/ml; P < 0.0005), and increased further with bosentan therapy (1.46 vs. 3.05 pg/ml; t-test P < 0.05). Bosentan was well tolerated with no significant drug-related serious adverse events and long-term outcomes were favourable compared with historic cases. Three patients developed rebound hypertension on withdrawal of bosentan and one appeared to further benefit from maintenance therapy. CONCLUSION: Upregulation of ET-1 ligand axis suggests that ET-1 receptor blockade is logical and treatment with bosentan appears to be safe in SRC. Future studies to assess therapeutic benefit and compare selective or non-selective receptor antagonists are justified.


Assuntos
Injúria Renal Aguda/tratamento farmacológico , Anti-Hipertensivos/uso terapêutico , Endotelina-1/sangue , Receptor de Endotelina A/metabolismo , Escleroderma Sistêmico/tratamento farmacológico , Sulfonamidas/uso terapêutico , Injúria Renal Aguda/etiologia , Adulto , Idoso , Bosentana , Estudos de Casos e Controles , Feminino , Humanos , Hipertensão/tratamento farmacológico , Hipertensão/etiologia , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Escleroderma Sistêmico/complicações , Sulfonamidas/metabolismo , Resultado do Tratamento
9.
Neurosci Lett ; 530(1): 41-6, 2012 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-23041043

RESUMO

Hippocampal CA1 neurons are particularly sensitive to ischemic damage, such as experienced following cardiac arrest and cardiopulmonary resuscitation. In recent years transient receptor potential M2 (TRPM2) channels have been identified as mediators of ischemic damage. We previously demonstrated that neuroprotective strategies targeting TRPM2 channels preferentially protect male cortical neurons from ischemic injury both in vitro and in vivo. It is important to determine the role of TRPM2 in ischemic injury of hippocampal neurons as this population of neurons are particularly sensitive to ischemic injury and are therapeutic targets. Here we report significantly decreased neuronal cell death following in vitro ischemia preferentially in male hippocampal neurons using TRPM2 inhibitors or knockdown of TRPM2 expression. Electrophysiological characterization of sex-stratified cultures shows similar levels of functional TRPM2 channel expression in male and female hippocampal neurons under basal conditions. In contrast, recordings made during reperfusion following in vitro ischemia revealed that TRPM2 channels are activated only in male neurons, resulting in rapid and complete depolarization. These findings provide strong evidence for TRPM2 as a target for protection against cerebral ischemia in male brain and helps define a molecular cell death pathway that is differentially engaged in male and female neurons.


Assuntos
Morte Celular/fisiologia , Hipocampo/patologia , Isquemia/fisiopatologia , Caracteres Sexuais , Canais de Cátion TRPM/fisiologia , Potenciais de Ação/fisiologia , Animais , Feminino , Técnicas de Silenciamento de Genes , Glucose/farmacologia , Isquemia/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo/fisiologia , Oxigênio/farmacologia , Técnicas de Patch-Clamp , Gravidez , Cultura Primária de Células , Canais de Cátion TRPM/genética
10.
Transl Stroke Res ; 3(4): 500-507, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23483801

RESUMO

Female sex steroids, particularly estrogens, contribute to the sexually dimorphic response observed in cerebral ischemic outcome, with females being relatively protected compared to males. Using a mouse model of cardiac arrest and cardiopulmonary resuscitation (CA/CPR), we previously demonstrated that estrogen neuroprotection is mediated in part by the estrogen receptor ß, with no involvement of estrogen receptor α. In this study we examined the neuroprotective effect of the novel estrogen receptor, G-protein coupled estrogen receptor 1 (GPER1/GPR30). Male mice administered the GPR30 agonist G1 exhibited significantly reduced neuronal injury in the hippocampal CA1 region and striatum. The magnitude of neuroprotection observed in G1 treated mice was indistinguishable from estrogen treated mice, implicating GPR30 in estrogen neuroprotection. Real-time quantitative RT-PCR indicates that G1 treatment increases expression of the neuroprotective ion channel, small conductance calcium-activated potassium channel 2. We conclude that GPR30 agonists show promise in reducing brain injury following global cerebral ischemia.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA