Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Food Chem Toxicol ; 190: 114789, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38844066

RESUMO

The safety assessments for chemicals targeted for use or expected to be exposed to specific life stages, including infancy, childhood, pregnancy and lactation, and geriatrics, need to account for extrapolation of data from healthy adults to these populations to assess their human health risk. However, often adequate and relevant toxicity or pharmacokinetic (PK) data of chemicals in specific life stages are not available. For such chemicals, New Approach Methodologies (NAMs), such as physiologically based pharmacokinetic (PBPK) modeling, biologically based dose response (BBDR) modeling, in vitro to in vivo extrapolation (IVIVE), etc. can be used to understand the variability of exposure and effects of chemicals in specific life stages and assess their associated risk. A life stage specific PBPK model incorporates the physiological and biochemical changes associated with each life stage and simulates their impact on the absorption, distribution, metabolism, and elimination (ADME) of these chemicals. In our review, we summarize the parameterization of life stage models based on New Approach Methodologies (NAMs) and discuss case studies that highlight the utility of a life stage based PBPK modeling for risk assessment. In addition, we discuss the utility of artificial intelligence (AI)/machine learning (ML) and other computational models, such as those based on in vitro data, as tools for estimation of relevant physiological or physicochemical parameters and selection of model. We also discuss existing gaps in the available toxicological datasets and current challenges that need to be overcome to expand the utility of NAMs for life stage-specific chemical risk assessment.


Assuntos
Modelos Biológicos , Medição de Risco/métodos , Humanos , Animais , Estágios do Ciclo de Vida/efeitos dos fármacos , Feminino
2.
Food Chem Toxicol ; 183: 114333, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38061571

RESUMO

The 6:2 fluorotelomer alcohol (6:2 FTOH) is a common impurity in per- and polyfluoroalkyl substances (PFASs) used in many applications. Our previous toxicokinetic (TK) evaluation of 6:2 FTOH calculated times to steady state (tss) of one of its metabolites, 5:3 fluorotelomer carboxylic acid (5:3A), in the plasma and tissues of up to a year after oral exposure to rats. Our current work further elucidated the TK of 5:3A and other metabolites of 6:2 FTOH in pregnant and nonpregnant rats after repeated oral exposure and examined the role of renal transporters in the biopersistence of 5:3A. The tss values for 5:3A in serum and tissues of adult nonpregnant animals ranged from 150 days to over a year. 4:3 fluorotelomer carboxylic acid (4:3A) was an additional potentially-biopersistent metabolite. 5:3A was the major metabolite of 6:2 FTOH in serum of pregnant dams and fetuses at each time interval. 5:3A was not a substrate for renal transporters in a human kidney cell line in vitro, indicating that renal reuptake of 5:3A is unlikely contribute to its biopersistence. Further research is needed to identify the underlying processes and evaluate the impact of these 6:2 FTOH metabolites on human health.


Assuntos
Fluorocarbonos , Ratos , Humanos , Animais , Gravidez , Feminino , Toxicocinética , Fluorocarbonos/toxicidade , Fluorocarbonos/química , Transporte Biológico , Ácidos Carboxílicos
4.
Toxics ; 10(5)2022 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-35622645

RESUMO

During the past few decades, the science of toxicology has been undergoing a transformation from observational to predictive science. New approach methodologies (NAMs), including in vitro assays, in silico models, read-across, and in vitro to in vivo extrapolation (IVIVE), are being developed to reduce, refine, or replace whole animal testing, encouraging the judicious use of time and resources. Some of these methods have advanced past the exploratory research stage and are beginning to gain acceptance for the risk assessment of chemicals. A review of the recent literature reveals a burst of IVIVE publications over the past decade. In this review, we propose operational definitions for IVIVE, present literature examples for several common toxicity endpoints, and highlight their implications in decision-making processes across various federal agencies, as well as international organizations, including those in the European Union (EU). The current challenges and future needs are also summarized for IVIVE. In addition to refining and reducing the number of animals in traditional toxicity testing protocols and being used for prioritizing chemical testing, the goal to use IVIVE to facilitate the replacement of animal models can be achieved through their continued evolution and development, including a strategic plan to qualify IVIVE methods for regulatory acceptance.

5.
Biomolecules ; 13(1)2022 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-36671390

RESUMO

The safety evaluation of oral exposure to substances, such as food ingredients, additives, and their constituents, relies primarily on a careful evaluation and analysis of data from oral toxicity studies. When relevant oral toxicity studies are unavailable or may have significant data gaps that make them inadequate for use in safety evaluations, data from non-oral toxicity studies in animals, such as studies on inhalation, dermal exposure, etc., might be used in support of or in place of oral toxicity studies through route-to-route (R-t-R) extrapolation. R-t-R extrapolation is applied on a case-by-case basis as it requires attention to and comparison of substance-specific toxicokinetic (TK) and toxicodynamic (TD) data for oral and non-oral exposure routes. This article provides a commentary on the utility of R-t-R extrapolation to assess the safety of oral exposure to substances, with an emphasis on the relevance of TK and systemic toxicity data.


Assuntos
Administração por Inalação , Segurança , Animais
6.
Toxicol Appl Pharmacol ; 422: 115531, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33933458

RESUMO

Perfluoropolyethers, also known as ether-PFAS, are linear or branched alkyl ether polymers, where the substituent hydrogens on the carbon atoms in the chain have been fully replaced by fluorine atoms. Some of these molecules may have a carboxylate functional group attached to one of the terminal carbon atoms to form an ether-PFAS carboxylate. Perfluoropolyethers are used as processing aids in the manufacture of various types of perfluorinated polymeric materials which are used in a variety of consumer applications. Although the physicochemical and toxicological properties of certain perfluoropolyether compounds have been extensively studied, data are relatively sparse for some members of this class of compounds. Moreover, the physicochemical, toxicokinetic, and toxicological properties of ether-PFAS as a class have not been elucidated in previous comprehensive review articles. This article reviews the nomenclature and uses of ether-PFAS and compares the physicochemical properties, toxicokinetic characteristics, apical effects in toxicological studies, and dose-response profiles across four specific ether-PFAS compounds. This comparison, including a description of identified data gaps should help to inform the design of studies to further elucidate the characteristics of ether-PFAS and to propose potential read-across assessment strategies for members of this class.


Assuntos
Poluentes Ambientais/toxicidade , Éteres/toxicidade , Fluorocarbonos/toxicidade , Testes de Toxicidade , Animais , Relação Dose-Resposta a Droga , Poluentes Ambientais/química , Éteres/química , Fluorocarbonos/química , Humanos , Estrutura Molecular , Medição de Risco , Relação Estrutura-Atividade , Toxicocinética
9.
Food Chem Toxicol ; 138: 111210, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32087313

RESUMO

6:2 Fluorotelomer alcohol (6:2 FTOH) is a short-chain polyfluoroalkyl substance (PFAS) in polymeric PFAS used in fast food packaging and stain- and water-resistant textiles and may be degradation products of some components of aqueous film-forming foams (AFFF). The general population is exposed to 6:2 FTOH by inhalation of evaporates from treated surfaces or ambient concentrations in air, ingestion of indoor dust, or ingestion of food packaged in materials containing PFAS. Although exposure to 6:2 FTOH is pervasive, little is known concerning human health effects of this compound. Some published risk assessments have assumed that perfluorohexanoic acid (PFHxA), a metabolite of 6:2 FTOH, adequately models the human health effects of 6:2 FTOH. Recently identified studies conducted with 6:2 FTOH and its metabolite, 5:3 acid, have provided information that enables comparison of the toxicological profiles of PFHxA and 6:2 FTOH. This article summarizes a comparative analysis of the toxicological effects of PFHxA and 6:2 FTOH in rodents to determine whether data for PFHxA adequately models potential hazards of 6:2 FTOH exposure. Our analysis demonstrates that 6:2 FTOH is significantly more toxic than PFHxA. Use of toxicological studies conducted with PFHxA to assess 6:2 FTOH exposure may significantly underestimate human health risk.


Assuntos
Álcoois/toxicidade , Fluorocarbonos/toxicidade , Toxicologia , Álcoois/química , Animais , Caproatos , Bases de Dados Factuais , Fluorocarbonos/química , Humanos , Medição de Risco
10.
Toxicol Appl Pharmacol ; 388: 114878, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31923437

RESUMO

Our previous report on pharmacokinetic (PK) evaluation of 6:2 fluorotelomer alcohol (6:2 FTOH) examined the biopersistence potential of its metabolites based on data published from single inhalation and occupational 6:2 FTOH exposure studies. We calculated internal exposure estimates of three key metabolites of 6:2 FTOH, of which 5:3 fluorotelomer carboxylic acid (5:3 acid) had the highest internal exposure and the slowest clearance. No oral repeated 6:2 FTOH exposure data were available at the time to fully characterize the biopersistence potential of the metabolite 5:3 acid. We recently received additional data on 6:2 FTOH and 5:3 acid, which included a 90-day toxicokinetic study report on repeated oral 6:2 FTOH exposure to rats. We reviewed the study and analyzed the reported 5:3 acid concentrations in plasma, liver, and fat using one-compartment PK modeling and calculated elimination rate constants (kel), elimination half-lives (t1/2) and times to steady state (tss) of 5:3 acid at three 6:2 FTOH doses. Our results showed that tss of 5:3 acid in plasma and evaluated tissues were approximately close to 1 year, such that the majority of highest values were observed at the lowest 6:2 FTOH dose, indicating its association with the biopersistence of 6:2 FTOH. The results of our PK analysis are the first to characterize biopersistence potential of the 5:3 acid after repeated oral exposure to the parent compound 6:2 FTOH based on steady state PK parameters, and therefore, may have an impact on future study designs when conducting toxicity assays for such compounds.


Assuntos
Polímeros de Fluorcarboneto/farmacocinética , Tecido Adiposo/química , Tecido Adiposo/efeitos dos fármacos , Administração Oral , Animais , Feminino , Polímeros de Fluorcarboneto/administração & dosagem , Polímeros de Fluorcarboneto/análise , Polímeros de Fluorcarboneto/toxicidade , Meia-Vida , Fígado/química , Fígado/efeitos dos fármacos , Masculino , Taxa de Depuração Metabólica , Ratos , Projetos de Pesquisa , Fatores de Tempo , Testes de Toxicidade Crônica/métodos
11.
Toxicol Appl Pharmacol ; 382: 114759, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31521730

RESUMO

The use of toxicokinetic (TK) data is becoming more prevalent in the evaluation of food ingredient safety as more TK information is being incorporated in safety data packages. Data demonstrating "1) the extent of absorption, 2) tissue distribution, 3) pathways and rates of metabolism, and 4) rate(s) of elimination" of food ingredients and their metabolites of intermediate and high toxicological potential may be useful for planning and designing toxicity studies, selecting doses for toxicity studies, addressing species differences, and understanding the potential modes of action to evaluate their safety. TK data reported in the literature or generated from mechanistic TK studies can be analyzed using mathematical methods, including compartment and noncompartment TK methods, whose predictions can enhance interpretation of observed effects. Because of recent advancements, several approaches have been developed to improve sensitivity of analyses of available TK data and reduce uncertainty for evaluating safety of food ingredients. An example of advanced TK methods is physiologically-based TK (PBTK) modeling that incorporates physiological/biochemical parameters into a TK framework to predict internal exposure. In this review, we discuss the utility of some TK methods and explore their relevance and potential value for food ingredient safety evaluation. We also describe the strengths and limitations of these TK methods and discuss current challenges and opportunities for expanding their application for evaluating safety of food ingredients. This review represents a state of science report, and not a guidance document, on the utility and relevance of TK methods for the safety evaluation of food ingredients.


Assuntos
Ingredientes de Alimentos/toxicidade , Inocuidade dos Alimentos/métodos , Toxicocinética , Animais , Humanos , Medição de Risco/métodos , Distribuição Tecidual/efeitos dos fármacos , Distribuição Tecidual/fisiologia
12.
Food Chem Toxicol ; 112: 375-382, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29331735

RESUMO

Polyfluorinated compounds (PFCs) are authorized for use as greaseproofing agents in food contact paper. As C8-PFCs (8-carbons) are known to accumulate in tissues, shorter-chain C6-PFCs (6-carbons) have replaced C8-PFCs in many food contact applications. However, the potential of C6-PFCs for human biopersistence has not been fully evaluated. For the first time, we provide internal exposure estimates to key metabolites of 6:2 fluorotelomer alcohol (6:2 FTOH), a monomeric component of C6-PFCs, to extend our understanding of exposure beyond estimates of external exposure. Pharmacokinetic data from published rat and human studies on 6:2 FTOH were used to estimate clearance and area under the curve (AUC) for its metabolites: 5:3 fluorotelomer carboxylic acid (5:3 A), perfluorohexanoic acid (PFHxA) and perfluoroheptanoic acid (PFHpA). Internal exposure to 5:3 A was the highest of evaluated metabolites across species and it had the slowest clearance. Additionally, 5:3 A clearance decreased with increasing 6:2 FTOH exposure. Our analysis provides insight into association of increased internal 5:3 A exposure with high biopersistence potential of 6:2 FTOH. Our results identify 5:3 A as an important biomarker of internal 6:2 FTOH exposure for use in biomonitoring studies, and are potentially useful for toxicological assessment of chronic dietary 6:2 FTOH exposure.


Assuntos
Fluorocarbonos/farmacocinética , Animais , Feminino , Fluorocarbonos/sangue , Fluorocarbonos/química , Humanos , Masculino , Estrutura Molecular , Ratos
13.
J Neuroinflammation ; 13(1): 299, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27903275

RESUMO

BACKGROUND: Traumatic brain injury (TBI) patients in military settings can be exposed to prolonged periods of hypobaria (HB) during aeromedical evacuation. Hypobaric exposure, even with supplemental oxygen to prevent hypoxia, worsens outcome after experimental TBI, in part by increasing neuroinflammation. Cell cycle activation (CCA) after TBI has been implicated as a mechanism contributing to both post-traumatic cell death and neuroinflammation. Here, we examined whether hypobaric exposure in rats subjected to TBI increases CCA and microglial activation in the brain, as compared to TBI alone, and to evaluate the ability of a cyclin-dependent kinase (CDK) inhibitor (CR8) to reduce such changes and improve behavioral outcomes. METHODS: Adult male Sprague Dawley rats were subjected to fluid percussion-induced injury, and HB exposure was performed at 6 h after TBI. Western blot and immunohistochemistry (IHC) were used to assess cell cycle-related protein expression and inflammation at 1 and 30 days after injury. CR8 was administered intraperitoneally at 3 h post-injury; chronic functional recovery and histological changes were assessed. RESULTS: Post-traumatic hypobaric exposure increased upregulation of cell cycle-related proteins (cyclin D1, proliferating cell nuclear antigen, and CDK4) and microglial/macrophage activation in the ipsilateral cortex at day 1 post-injury as compared to TBI alone. Increased immunoreactivity of cell cycle proteins, as well as numbers of Iba-1+ and GFAP+ cells in both the ipsilateral cortex and hippocampus were found at day 30 post-injury. TBI/HB significantly increased the numbers of NADPH oxidase 2 (gp91phox) enzyme-expressing cells that were co-localized with Iba-1+. Each of these changes was significantly reduced by the administration of CR8. Unbiased stereological assessment showed significantly decreased numbers of microglia displaying the highly activated phenotype in the ipsilateral cortex of TBI/HB/CR8 rats compared with TBI/HB/Veh rats. Moreover, treatment with this CDK inhibitor also significantly improved spatial and retention memory and reduced lesion volume and hippocampal neuronal cell loss. CONCLUSIONS: HB exposure following TBI increases CCA, neuroinflammation, and associated neuronal cell loss. These changes and post-traumatic cognitive deficits are reduced by CDK inhibition; such drugs may therefore serve to protect TBI patients requiring aeromedical evacuation.


Assuntos
Pressão Atmosférica , Lesões Encefálicas Traumáticas/metabolismo , Encéfalo/metabolismo , Ciclo Celular/fisiologia , Transtornos Cognitivos/metabolismo , Mediadores da Inflamação/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/imunologia , Ciclo Celular/efeitos dos fármacos , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/imunologia , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/metabolismo , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/imunologia , Masculino , Neurônios/efeitos dos fármacos , Neurônios/imunologia , Neurônios/metabolismo , Purinas/farmacologia , Piridinas/farmacologia , Ratos , Ratos Sprague-Dawley
14.
J Neurotrauma ; 33(14): 1292-302, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-26593382

RESUMO

Aeromedical evacuation, an important component in the care of many patients with traumatic brain injury (TBI), particularly in war zones, exposes them to prolonged periods of hypobaria. The effects of such exposure on pathophysiological changes and outcome after TBI are largely unexplored. The objective of this study was to investigate whether prolonged hypobaria in rats subjected to TBI alters behavioral and histological outcomes. Adult male Sprague-Dawley rats underwent fluid percussion induced injury at 1.5-1.9 atmospheres of pressure. The effects of hypobaric exposure (6 h duration; equivalent to 0.75 atmospheres) at 6, 24, and 72 h, or 7 days after TBI were evaluated with regard to sensorimotor, cognitive, and histological changes. Additional groups were evaluated to determine the effects of two hypobaric exposures after TBI, representing primary simulated aeromedical evacuation (6 h duration at 24 h after injury) and secondary evacuation (10 h duration at 72 h after injury), as well as the effects of 100% inspired oxygen concentrations during simulated evacuation. Hypobaric exposure up to 7 days after injury significantly worsened cognitive deficits, hippocampal neuronal loss, and microglial/astrocyte activation in comparison with injured controls not exposed to hypobaria. Hyperoxia during hypobaric exposure or two exposures to prolonged hypobaric conditions further exacerbated spatial memory deficits. These findings indicate that exposure to prolonged hypobaria up to 7 days after TBI, even while maintaining physiological oxygen concentration, worsens long-term cognitive function and neuroinflammation. Multiple exposures or use of 100% oxygen further exacerbates these pathophysiological effects.


Assuntos
Pressão do Ar , Comportamento Animal/fisiologia , Lesões Encefálicas Traumáticas , Disfunção Cognitiva , Hipocampo , Hiperóxia , Inflamação , Animais , Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/imunologia , Lesões Encefálicas Traumáticas/patologia , Lesões Encefálicas Traumáticas/fisiopatologia , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/imunologia , Disfunção Cognitiva/patologia , Disfunção Cognitiva/fisiopatologia , Modelos Animais de Doenças , Hipocampo/patologia , Inflamação/etiologia , Inflamação/imunologia , Masculino , Ratos , Ratos Sprague-Dawley , Memória Espacial/fisiologia
15.
J Cereb Blood Flow Metab ; 35(12): 2010-20, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26154869

RESUMO

Neuroinflammation following traumatic brain injury (TBI) is increasingly recognized to contribute to chronic tissue loss and neurologic dysfunction. Circulating levels of S100B increase after TBI and have been used as a biomarker. S100B is produced by activated astrocytes and can promote microglial activation; signaling by S100B through interaction with the multiligand advanced glycation end product-specific receptor (AGER) has been implicated in brain injury and microglial activation during chronic neurodegeneration. We examined the effects of S100B inhibition in a controlled cortical impact model, using S100B knockout mice or administration of neutralizing S100B antibody. Both interventions significantly reduced TBI-induced lesion volume, improved retention memory function, and attenuated microglial activation. The neutralizing antibody also significantly reduced sensorimotor deficits and improved neuronal survival in the cortex. However, S100B did not alter microglial activation in BV2 cells or primary microglial cultures stimulated by lipopolysaccharide or interferon gamma. Further, proximity ligation assays did not support direct interaction in the brain between S100B and AGER following TBI. Future studies are needed to elucidate specific pathways underlying S100B-mediated neuroinflammatory actions after TBI. Our results strongly implicate S100B in TBI-induced neuroinflammation, cell loss, and neurologic dysfunction, thereby indicating that it is a potential therapeutic target for TBI.


Assuntos
Comportamento Animal , Lesões Encefálicas/patologia , Lesões Encefálicas/psicologia , Encéfalo/patologia , Subunidade beta da Proteína Ligante de Cálcio S100/antagonistas & inibidores , Subunidade beta da Proteína Ligante de Cálcio S100/genética , Animais , Anticorpos Neutralizantes/farmacologia , Linhagem Celular , Inflamação/patologia , Interferon gama/farmacologia , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos , Masculino , Memória , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia , Equilíbrio Postural , Receptor para Produtos Finais de Glicação Avançada/genética , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Reconhecimento Psicológico
16.
J Neurotrauma ; 32(5): 289-96, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25242371

RESUMO

Traumatic brain injury (TBI) can cause sleep-wake disturbances and excessive daytime sleepiness. The pathobiology of sleep disorders in TBI, however, is not well understood, and animal models have been underused in studying such changes and potential underlying mechanisms. We used the rat lateral fluid percussion (LFP) model to analyze sleep-wake patterns as a function of time after injury. Rapid-eye movement (REM) sleep, non-REM (NREM) sleep, and wake bouts during light and dark phases were measured with electroencephalography and electromyography at an early as well as chronic time points after LFP. Moderate TBI caused disturbances in the ability to maintain consolidated wake bouts during the active phase and chronic loss of wakefulness. Further, TBI resulted in cognitive impairments and depressive-like symptoms, and reduced the number of orexin-A-positive neurons in the lateral hypothalamus.


Assuntos
Lesões Encefálicas/complicações , Transtornos do Sono do Ritmo Circadiano/etiologia , Transtornos do Sono do Ritmo Circadiano/fisiopatologia , Vigília/fisiologia , Animais , Lesões Encefálicas/metabolismo , Lesões Encefálicas/fisiopatologia , Modelos Animais de Doenças , Eletroencefalografia , Eletromiografia , Hipotálamo/metabolismo , Imuno-Histoquímica , Masculino , Orexinas/análise , Orexinas/biossíntese , Ratos , Ratos Sprague-Dawley , Transtornos do Sono do Ritmo Circadiano/metabolismo
17.
Neural Regen Res ; 9(17): 1578-80, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25368642

RESUMO

Traumatic brain injury induces secondary injury that contributes to neuroinflammation, neuronal loss, and neurological dysfunction. One important injury mechanism is cell cycle activation which causes neuronal apoptosis and glial activation. The neuroprotective effects of both non-selective (Flavopiridol) and selective (Roscovitine and CR-8) cyclin-dependent kinase inhibitors have been shown across multiple experimental traumatic brain injury models and species. Cyclin-dependent kinaseinhibitors, administered as a single systemic dose up to 24 hours after traumatic brain injury, provide strong neuroprotection-reducing neuronal cell death, neuroinflammation and neurological dysfunction. Given their effectiveness and long therapeutic window, cyclin-dependent kinase inhibitors appear to be promising candidates for clinical traumatic brain injury trials.

18.
J Cereb Blood Flow Metab ; 34(7): 1223-32, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24756076

RESUMO

Repeated mild traumatic brain injury (mTBI) can cause sustained cognitive and psychiatric changes, as well as neurodegeneration, but the underlying mechanisms remain unclear. We examined histologic, neurophysiological, and cognitive changes after single or repeated (three injuries) mTBI using the rat lateral fluid percussion (LFP) model. Repeated mTBI caused substantial neuronal cell loss and significantly increased numbers of activated microglia in both ipsilateral and contralateral hippocampus on post-injury day (PID) 28. Long-term potentiation (LTP) could not be induced on PID 28 after repeated mTBI in ex vivo hippocampal slices from either hemisphere. N-Methyl-D-aspartate (NMDA) receptor-mediated responses were significantly attenuated after repeated mTBI, with no significant changes in α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor-mediated responses. Long-term potentiation was elicited in slices after single mTBI, with potentiation significantly increased in ipsilateral versus contralateral hippocampus. After repeated mTBI, rats displayed cognitive impairments in the Morris water maze (MWM) and novel object recognition (NOR) tests. Thus, repeated mTBI causes deficits in the hippocampal function and changes in excitatory synaptic neurotransmission, which are associated with chronic neuroinflammation and neurodegeneration.


Assuntos
Lesões Encefálicas/fisiopatologia , Transtornos Cognitivos/fisiopatologia , Hipocampo/fisiopatologia , Inflamação/fisiopatologia , Potenciação de Longa Duração/fisiologia , Animais , Lesões Encefálicas/complicações , Transtornos Cognitivos/etiologia , Modelos Animais de Doenças , Lateralidade Funcional/fisiologia , Imuno-Histoquímica , Inflamação/etiologia , Masculino , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Transmissão Sináptica/fisiologia
19.
J Neurotrauma ; 31(8): 758-72, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24476502

RESUMO

Traumatic brain injury (TBI) causes neuronal cell death as well as microglial activation and related neurotoxicity that contribute to subsequent neurological dysfunction. Poly (ADP-ribose) polymerase (PARP-1) induces neuronal cell death through activation of caspase-independent mechanisms, including release of apoptosis inducing factor (AIF), and microglial activation. Administration of PJ34, a selective PARP-1 inhibitor, reduced cell death of primary cortical neurons exposed to N-Methyl-N'-Nitro-N-Nitrosoguanidine (MNNG), a potent inducer of AIF-dependent cell death. PJ34 also attenuated lipopolysaccharide and interferon-γ-induced activation of BV2 or primary microglia, limiting NF-κB activity and iNOS expression as well as decreasing generation of reactive oxygen species and TNFα. Systemic administration of PJ34 starting as late as 24 h after controlled cortical impact resulted in improved motor function recovery in mice with TBI. Stereological analysis demonstrated that PJ34 treatment reduced the lesion volume, attenuated neuronal cell loss in the cortex and thalamus, and reduced microglial activation in the TBI cortex. PJ34 treatment did not improve cognitive performance in a Morris water maze test or reduce neuronal cell loss in the hippocampus. Overall, our data indicate that PJ34 has a significant, albeit selective, neuroprotective effect after experimental TBI, and its therapeutic effect may be from multipotential actions on neuronal cell death and neuroinflammatory pathways.


Assuntos
Lesões Encefálicas/patologia , Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fenantrenos/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases , Animais , Western Blotting , Lesões Encefálicas/enzimologia , Morte Celular/efeitos dos fármacos , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/patologia , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Poli(ADP-Ribose) Polimerase-1 , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos
20.
Int J Mol Sci ; 15(1): 1216-36, 2014 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-24445258

RESUMO

Traumatic brain injury (TBI) induces secondary biochemical changes that contribute to delayed neuroinflammation, neuronal cell death, and neurological dysfunction. Attenuating such secondary injury has provided the conceptual basis for neuroprotective treatments. Despite strong experimental data, more than 30 clinical trials of neuroprotection in TBI patients have failed. In part, these failures likely reflect methodological differences between the clinical and animal studies, as well as inadequate pre-clinical evaluation and/or trial design problems. However, recent changes in experimental approach and advances in clinical trial methodology have raised the potential for successful clinical translation. Here we critically analyze the current limitations and translational opportunities for developing successful neuroprotective therapies for TBI.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Pesquisa Translacional Biomédica/normas , Animais , Ensaios Clínicos como Assunto , Humanos , Pesquisa Translacional Biomédica/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA