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










Base de dados
Intervalo de ano de publicação
1.
J Am Assoc Lab Anim Sci ; 52(1): 17-21, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23562028

RESUMO

The use of a murine nonsurgical embryo transfer (NSET) device had been described previously for the transfer of blastocysts, morulae, DNA-microinjected embryos, and embryonic stem cell-containing embryos to create genetically modified mice. However, physiologic effects of the NSET device and traditional surgical methods had not been compared directly. Here we used electrocardiography and fecal corticosterone levels to monitor pseudopregnant mice that underwent anesthesia only, the NSET procedure with or without anesthesia, or surgery. These procedures were performed without the use of actual embryos, to focus on effects of the procedures themselves rather than on any physiologic effects due to the deposition of embryos. As compared with surgery and anesthesia, the NSET procedure was associated with less fluctuation in cardiac rhythm and lower levels of the stress biomarker fecal corticosterone. These results indicate that use of the NSET device avoids these physi- ological perturbations as well as other disadvantages of surgery (for example, postoperative pain and need for postoperative analgesia) and therefore provides a valuable refinement of existing mouse embryo transfer procedures.


Assuntos
Transferência Embrionária/veterinária , Camundongos Transgênicos/embriologia , Anestesia Geral/veterinária , Bem-Estar do Animal , Animais , Blastocisto , Transferência Embrionária/métodos , Feminino , Camundongos , Mórula , Gravidez
2.
J Neurochem ; 117(3): 579-88, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21361959

RESUMO

Free radical-induced lipid peroxidation (LP) is critical in the evolution of secondary injury following traumatic brain injury (TBI). Previous studies in our laboratory demonstrated that U-83836E, a potent LP inhibitor, can reduce post-TBI LP along with an improved maintenance of mouse cortical mitochondrial bioenergetics and calcium (Ca(2+)) buffering following severe (1.0 mm; 3.5 m/s) controlled cortical impact TBI (CCI-TBI). Based upon this preservation of a major Ca(2+) homeostatic mechanism, we have now performed dose-response and therapeutic window analyses of the ability of U-83836E to reduce post-traumatic calpain-mediated cytoskeletal (α-spectrin) proteolysis in ipsilateral cortical homogenates at its 24 h post-TBI peak. In the dose-response analysis, mice were treated with a single i.v. dose of vehicle or U-83836E (0.1, 0.3, 1.3, 3.0, 10.0 or 30.0 mg/kg) at 15 min after injury. U-83836E produced a dose-related attenuation of α-spectrin degradation with the maximal decrease being achieved at 3.0 mg/kg. Next, the therapeutic window was tested by delaying the single 3 mg/kg i.v. dose from 15 min post-injury out to 1, 3, 6 or 12 h. No reduction in α-spectrin degradation was observed when the treatment delay was 1 h or longer. However, in a third experiment, we re-examined the window with repeated U-83836E dosing (3.0 mg/kg i.v. followed by 10 mg/kg i.p. maintenance doses at 1 and 3 h after the initial i.v. dose) which significantly reduced 24 h α-α-spectrin degradation even when treatment initiation was withheld until 12 h post-TBI. These results demonstrate the relationship between post-TBI LP, disruptions in neuronal Ca(2+) homeostasis and calpain-mediated cytoskeletal damage.


Assuntos
Lesões Encefálicas , Calpaína/farmacologia , Cromanos/uso terapêutico , Citoesqueleto/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Piperazinas/uso terapêutico , Animais , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Cromanos/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Masculino , Camundongos , Fármacos Neuroprotetores/farmacologia , Piperazinas/farmacologia , Fatores de Tempo
3.
J Neurotrauma ; 27(12): 2233-43, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20874056

RESUMO

The cytoskeletal and neuronal protective effects of early treatment with the blood-brain barrier- and cell-permeable calpain inhibitor MDL-28170 was examined in the controlled cortical impact (CCI) traumatic brain injury (TBI) model in male CF-1 mice. This was preceded by a dose-response and pharmacodynamic evaluation of IV or IP doses of MDL-28170 with regard to ex vivo inhibition of calpain 2 activity in harvested brain homogenates. From these data, we tested the effects of an optimized MDL-28170 dosing regimen on calpain-mediated degradation of the neuronal cytoskeletal protein α-spectrin in cortical or hippocampal tissue of mice 24 h after CCI-TBI (1.0 mm depth, 3.5 m/sec velocity). With treatment initiated at 15 min post-TBI, α-spectrin degradation was significantly reduced by 40% in hippocampus and 44% in cortex. This effect was still observed with a 1-h but not a 3-h post-TBI delay. The cytoskeletal protection is most likely taking place in neurons surrounding the area of mainly necrotic degeneration, since MDL-28170 did not reduce hemispheric lesion volume as measured by the aminocupric silver staining method. This lack of effect on lesion volume has been seen with other calpain inhibitors, which suggests that pharmacological calpain inhibition by itself, while able to reduce axonal injury, may not be able to produce a measurable reduction in lesion volume. This is in contrast to certain other neuroprotective mechanistic approaches such as the mitochondrial protectant cyclosporine A, which produces at least a partial decrease in lesion volume in the same model. Accordingly, the combination of a calpain inhibitor with a compound such as cyclosporine A may be needed to achieve the optimal degree of post-TBI neuroprotection.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Calpaína/antagonistas & inibidores , Córtex Cerebral/efeitos dos fármacos , Dipeptídeos/uso terapêutico , Hipocampo/efeitos dos fármacos , Degeneração Neural/tratamento farmacológico , Análise de Variância , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Western Blotting , Lesões Encefálicas/patologia , Córtex Cerebral/lesões , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Dipeptídeos/farmacologia , Relação Dose-Resposta a Droga , Hipocampo/metabolismo , Hipocampo/patologia , Masculino , Camundongos , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Espectrina/metabolismo
4.
J Neurochem ; 114(1): 271-80, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20403083

RESUMO

Mitochondrial dysfunction after traumatic brain injury (TBI) is manifested by increased levels of oxidative damage, loss of respiratory functions and diminished ability to buffer cytosolic calcium. This study investigated the detrimental effects of lipid peroxyl radicals (LOO(*)) and lipid peroxidation (LP) in brain mitochondria after TBI by examining the protective effects of U-83836E, a potent and selective scavenger of LOO(*) radicals. Male CF1 mice were subjected to severe controlled cortical impact TBI (CCI-TBI) and treated with either vehicle or U-83836E initiated i.v. at 15 min post-injury. Calcium (Ca(++)) buffering capacity and respiratory function were measured in isolated cortical mitochondrial samples taken from the ipsilateral hemisphere at 3 and 12 h post-TBI, respectively. In vehicle-treated injured mice, the cortical mitochondrial Ca(++) buffering capacity was reduced by 60% at 3 h post-injury (p < 0.001) and the respiratory control ratio was decreased by 27% at 12 h post-TBI, relative to sham, non-injured mice. U-83836E treatment significantly (p < 0.05) preserved Ca(++) buffering capacity and attenuated the reduction in respiratory control ratio values. Consistent with the functional effects of U-83836E being as a result of an attenuation of mitochondrial oxidative damage, the compound significantly (p < 0.001) reduced LP-generated 4-hydroxynonenal levels in both cortical homogenates and mitochondria at both 3 and 12 h post-TBI. Unexpectedly, U-83836E also reduced peroxynitrite-generated 3-nitrotyrosine in parallel with the reduction in 4-hydroxynonenal. The results demonstrate that LOO(*) radicals contribute to secondary brain mitochondrial dysfunction after TBI by propagating LP and protein nitrative damage in cellular and mitochondrial membranes.


Assuntos
Lesões Encefálicas/metabolismo , Cromanos/farmacologia , Sequestradores de Radicais Livres/farmacologia , Peroxidação de Lipídeos/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Piperazinas/farmacologia , Animais , Cálcio/metabolismo , Respiração Celular , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Cromanos/química , Radicais Livres/metabolismo , Masculino , Camundongos , Mitocôndrias/metabolismo , Fármacos Neuroprotetores/química , Estresse Oxidativo , Piperazinas/química , Estereoisomerismo
5.
J Neurotrauma ; 26(8): 1369-78, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19419247

RESUMO

The reactive nitrogen species peroxynitrite (PN) has been suggested to be an important mediator of the secondary oxidative damage that occurs following acute spinal cord injury (SCI). The PN decomposition products nitrogen dioxide (*NO(2)), hydroxyl radical (*OH), and carbonate radical (*CO(3)) are highly reactive with cellular lipids and proteins. In this immunohistochemical study, we examined the temporal (3, 24, and 72 h, and 1 and 2 weeks) and spatial relationships of PN-mediated oxidative damage in the contusion-injured rat thoracic spinal cord (IH device, 200 kdyn, T10) using 3-nitrotyrosine (3-NT), a marker for protein nitration by PN-derived *NO(2) and 4-hydroxynonenal (4-HNE), an indicator of lipid peroxidation (LP) initiated by any of the PN radicals. Minimal 3-NT or 4-HNE immunostaining was seen in sham, non-injured spinal cords. In contrast, both markers showed a substantial increase at 3 h post-injury at the epicenter, that extended throughout the gray matter and into the surrounding white matter. At 24 and 72 h, the oxidative damage expanded circumferentially to involve all but a small rim of white matter tissue at the injury site, and longitudinally as much as 6-9 mm in the rostral and caudal directions. The staining was observed in neuronal soma, axons, and microvessels. At all time points except 3 h, there was no significant difference in the mean rostral or caudal extent of 3-NT and 4-HNE staining. By 1, and more so at 2 weeks, the longitudinal extent of the oxidative damage staining was greatly decreased. The spatial and temporal overlap of 3-NT and 4-HNE staining supports the concept that PN is involved in both damage produced by lipid peroxidation and protein nitration, and that antioxidant agents that target PN or PN-derived radicals should be effective neuroprotectants for acute SCI if administered during the first post-injury hours.


Assuntos
Neurônios/metabolismo , Ácido Peroxinitroso/metabolismo , Traumatismos da Medula Espinal/metabolismo , Medula Espinal/metabolismo , Aldeídos/metabolismo , Animais , Feminino , Imuno-Histoquímica , Peroxidação de Lipídeos , Neurônios/patologia , Estresse Oxidativo , Ratos , Ratos Sprague-Dawley , Medula Espinal/patologia , Traumatismos da Medula Espinal/patologia , Vértebras Torácicas , Fatores de Tempo , Tirosina/análogos & derivados , Tirosina/metabolismo
6.
J Cereb Blood Flow Metab ; 29(1): 87-97, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18714331

RESUMO

Earlier experiments have shown that cyclosporin A (CsA) and its non-calcineurin inhibitory analog NIM811 attenuate mitochondrial dysfunction after experimental traumatic brain injury (TBI). Presently, we compared the neuroprotective effects of previously determined mitochondrial protective doses of CsA (20 mg/kg intraperitoneally) and NIM811 (10 mg/kg intraperitoneally) when administered at 15 mins postinjury in preventing cytoskeletal (alpha-spectrin) degradation, neurodegeneration, and neurological dysfunction after severe (1.0 mm) controlled cortical impact (CCI) TBI in mice. In a first set of experiments, we analyzed calpain-mediated alpha-spectrin proteolysis at 24 h postinjury. Both NIM811 and CsA significantly attenuated the increased alpha-spectrin breakdown products observed in vehicle-treated animals (P<0.005). In a second set of experiments, treatment of animals with either NIM811 or CsA at 15 mins and again at 24 h postinjury attenuated motor function impairment at 48 h and 7 days (P<0.005) and neurodegeneration at 7 days postinjury (P<0.0001). Delayed administration of NIM811 out to 12 h was still able to significantly reduce alpha-spectrin degradation. These results show that the neuroprotective mechanism of CsA involves maintenance of mitochondrial integrity and that calcineurin inhibition plays little or no role because the non-calcineurin inhibitory analog, NIM811, is as effective as CsA.


Assuntos
Lesões Encefálicas/prevenção & controle , Ciclosporina/química , Ciclosporina/farmacologia , Fármacos Neuroprotetores/farmacologia , Animais , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Calpaína/metabolismo , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/enzimologia , Imunossupressores/química , Imunossupressores/farmacologia , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Fármacos Neuroprotetores/química , Condicionamento Físico Animal , Espectrina/metabolismo
7.
J Neurosci Res ; 86(10): 2258-70, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18381764

RESUMO

Memory impairment is one of the most significant residual deficits following traumatic brain injury (TBI) and is among the most frequent complaints heard from patients and their relatives. It has been reported that the hippocampus is particularly vulnerable to TBI, which results in hippocampus-dependent cognitive impairment. There are different regions in the hippocampus, and each region is composed of different cell types, which might respond differently to TBI. However, regional and cell type-specific neuronal death following TBI is not well described. Here, we examined the distribution of degenerating neurons in the hippocampus of the mouse brain following controlled cortical impact (CCI) and found that the majority of degenerating neurons observed were in the dentate gyrus after moderate (0.5 mm cortical deformation) CCI-TBI. In contrast, there were only a few degenerating neurons observed in the hilus, and we did not observe any degenerating neurons in the CA3 or CA1 regions. Among those degenerating cells in the dentate gyrus, about 80% of them were found in the inner granular neuron layer. Analysis with cell type-specific markers showed that most of the degenerating neurons in the inner granular neuron layer are newborn immature neurons. Further quantitative analysis shows that the number of newborn immature neurons in the dentate gyrus is dramatically decreased in the ipsilateral hemisphere compared with the contralateral side. Collectively, our data demonstrate the selective death of newborn immature neurons in the hippocampal dentate gyrus following moderate injury with CCI in mice. This selective vulnerability of newborn immature dentate neurons may contribute to the persistent impairment of learning and memory post-TBI and provide an innovative target for neuroprotective treatment strategies.


Assuntos
Lesões Encefálicas/patologia , Giro Denteado/patologia , Degeneração Neural/patologia , Neurônios/patologia , Animais , Morte Celular , Lateralidade Funcional , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL
8.
J Cereb Blood Flow Metab ; 28(6): 1114-26, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18319733

RESUMO

We examined the ability of tempol, a catalytic scavenger of peroxynitrite (PN)-derived free radicals, to reduce cortical oxidative damage, mitochondrial dysfunction, calpain-mediated cytoskeletal (alpha-spectrin) degradation, and neurodegeneration, and to improve behavioral recovery after a severe (depth 1.0 mm), unilateral controlled cortical impact traumatic brain injury (CCI-TBI) in male CF-1 mice. Administration of a single 300 mg/kg intraperitoneal dose of tempol 15 mins after TBI produced a complete suppression of PN-mediated oxidative damage (3-nitrotyrosine, 3NT) in injured cortical tissue at 1 h after injury. Identical tempol dosing maintained respiratory function and attenuated 3NT in isolated cortical mitochondria at 12 h after injury, the peak of mitochondrial dysfunction. Multiple dosing with tempol (300 mg/kg intraperitoneally at 15 mins, 3, 6, 9, and 12 h) also suppressed alpha-spectrin degradation by 45% at its 24 h post-injury peak. The same dosing regimen improved 48 h motor function and produced a significant, but limited (17.4%, P<0.05), decrease in hemispheric neurodegeneration at 7 days. These results are consistent with a mechanistic link between PN-mediated oxidative damage to brain mitochondria, calpain-mediated proteolytic damage, and neurodegeneration. However, the modest neuroprotective effect of tempol suggests that multitarget combination strategies may be needed to interfere with posttraumatic secondary injury to a degree worthy of clinical translation.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , Óxidos N-Cíclicos/uso terapêutico , Radicais Livres/metabolismo , Ácido Peroxinitroso/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Calpaína/metabolismo , Catálise , Óxidos N-Cíclicos/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Masculino , Camundongos , Mitocôndrias/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espectrina/metabolismo , Marcadores de Spin
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...