Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Am J Respir Cell Mol Biol ; 61(3): 341-354, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30897338

RESUMEN

Hyperoxia plays a key role in the development of bronchopulmonary dysplasia (BPD), a chronic lung disease of preterm infants. Infants with BPD often have brain injury that leads to long-term neurodevelopmental impairment, but the underlying mechanisms that control BPD-induced neurodevelopmental impairment remain unclear. Our previous studies have shown that hyperoxia-induced BPD in rodents is associated with lung inflammasome activation. Here, we tested the hypothesis that hyperoxia-induced lung and brain injury is mediated by inflammasome activation, and that inhibition of caspase-1, a key component of the inflammasome, attenuates hyperoxia-induced lung and brain injury in neonatal mice. C57/BL6 mouse pups were randomized to receive daily intraperitoneal injections of Ac-YVAD-CMK, an irreversible caspase-1 inhibitor, or placebo during exposure to room air or hyperoxia (85% O2) for 10 days. We found that hyperoxia activated the NLRP1 inflammasome, increased production of mature IL-1ß, and upregulated expression of p30 gasdermin-D (GSDMD), the active form of GSDMD that is responsible for the programmed cell death mechanism of pyroptosis in both lung and brain tissue. Importantly, we show that inhibition of caspase-1 decreased IL-1ß activation and p30 GSDMD expression, and improved alveolar and vascular development in hyperoxia-exposed lungs. Moreover, caspase-1 inhibition also promoted cell proliferation in the subgranular zone and subventricular zone of hyperoxia-exposed brains, resulting in lessened atrophy of these zones. Thus, the inflammasome plays a critical role in hyperoxia-induced neonatal lung and brain injury, and targeting this pathway may be beneficial for the prevention of lung and brain injury in preterm infants.


Asunto(s)
Lesiones Encefálicas/metabolismo , Caspasa 1/metabolismo , Hiperoxia/metabolismo , Lesión Pulmonar/metabolismo , Animales , Animales Recién Nacidos , Proliferación Celular/fisiología , Humanos , Hipertensión Pulmonar/complicaciones , Recién Nacido , Inflamasomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Serpinas/farmacología , Proteínas Virales/farmacología
2.
J Neurotrauma ; 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38445369

RESUMEN

There is a growing body of evidence that the delivery of cell-derived exosomes normally involved in intracellular communication can reduce secondary injury mechanisms after brain and spinal cord injury and improve outcomes. Exosomes are nanometer-sized vesicles that are released by Schwann cells and may have neuroprotective effects by reducing post-traumatic inflammatory processes as well as promoting tissue healing and functional recovery. The purpose of this study was to evaluate the beneficial effects of human Schwann-cell exosomes (hSC-Exos) in a severe model of penetrating ballistic-like brain injury (PBBI) in rats and investigate effects on multiple outcomes. Human Schwann cell processing protocols followed Current Good Manufacturing Practices (cGMP) with exosome extraction and purification steps approved by the Food and Drug Administration for an expanded access single ALS patient Investigational New Drug. Anesthetized male Sprague-Dawley rats (280-350g) underwent PBBI surgery or Sham procedures and, starting 30 min after injury, received either a dose of hSC-Exos or phosphate-buffered saline through the jugular vein. At 48h after PBBI, flow cytometry analysis of cortical tissue revealed that hSC-Exos administration reduced the number of activated microglia and levels of caspase-1, a marker of inflammasome activation. Neuropathological analysis at 21 days showed that hSC-Exos treatment after PBBI significantly reduced overall contusion volume and decreased the frequency of Iba-1 positive activated and amoeboid microglia by immunocytochemical analysis. This study revealed that the systemic administration of hSC-Exos is neuroprotective in a model of severe TBI and reduces secondary inflammatory injury mechanisms and histopathological damage. The administration of hSC-Exos represents a clinically relevant cell-based therapy to limit the detrimental effects of neurotrauma or other progressive neurological injuries by impacting multiple pathophysiological events and promoting neurological recovery.

3.
Transl Res ; 257: 66-77, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36758791

RESUMEN

Traumatic Brain Injury (TBI) is a major cause of death and disability in the US and a recognized risk factor for the development of Alzheimer's disease (AD). The relationship between these conditions is not completely understood, but the conditions may share additive or synergistic pathological hallmarks that may serve as novel therapeutic targets. Heightened inflammasome signaling plays a critical role in the pathogenesis of central nervous system injury (CNS) and the release of apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) speck from neurons and activated microglia contribute significantly to TBI and AD pathology. This study investigated whether inflammasome signaling after TBI was augmented in AD and whether this signaling pathway impacted biochemical and neuropathological outcomes and overall cognitive function. Five-month-old, 3xTg mice and respective wild type controls were randomized and underwent moderate controlled cortical impact (CCI) injury or served as sham/uninjured controls. Animals were sacrificed at 1 hour, 1 day, or 1 week after TBI to assess acute pathology or at 12 weeks after assessing cognitive function. The ipsilateral cerebral cortex was processed for inflammasome protein expression by immunoblotting. Mice were evaluated for behavior by open field (3 days), novel object recognition (2 weeks), and Morris water maze (6 weeks) testing after TBI. There was a statistically significant increase in the expression of inflammasome signaling proteins Caspase-1, Caspase-8, ASC, and interleukin (IL)-1ß after TBI in both wild type and 3xTg animals. At 1-day post injury, significant increases in ASC and IL-1ß protein expression were measured in AD TBI mice compared to WT TBI. Behavioral testing showed that injured AD mice had altered cognitive function when compared to injured WT mice. Elevated Aß was seen in the ipsilateral cortex and hippocampus of sham and injured AD when compared to respective groups at 12 weeks post injury. Moreover, treatment of injured AD mice with IC100, an anti-ASC monoclonal antibody, inhibited the inflammasome, as evidenced by IL-1ß reduction in the injured cortex at 1-week post injury. These findings show that the inflammasome response is heightened in mice genetically predisposed to AD and suggests that AD may exacerbate TBI pathology. Thus, dampening inflammasome signaling may offer a novel approach for the treatment of AD and TBI.


Asunto(s)
Enfermedad de Alzheimer , Lesiones Traumáticas del Encéfalo , Ratones , Animales , Inflamasomas/metabolismo , Enfermedad de Alzheimer/genética , Predisposición Genética a la Enfermedad , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/genética , Apoptosis
4.
Front Aging Neurosci ; 14: 942717, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36062148

RESUMEN

Low-frequency whole-body vibration (WBV; 40 Hz), a low impact form of exercise, intervention for a month following moderate transient middle-cerebral artery occlusion (tMCAO) reduces infarct volume and improves motor function in reproductively senescent, middle-aged female rats. Since post-stroke cognitive decline remains a significant problem, the current study aims to investigate the efficacy of WBV in ameliorating post-tMCAO cognitive deficits and to determine the underlying putative mechanism(s) conferring benefits of WBV in middle-aged rats. Middle-aged rats of both sexes were randomly assigned to tMCAO (90 min) or sham surgery followed by exposure to either WBV (twice a day for 15 min each for 5 days a week over a month) or no WBV treatment groups. Following the last WBV treatment, rats were tested for hippocampus-dependent learning and memory using a water maze followed by harvesting brain and blood samples for histopathological and inflammatory marker analyses, respectively. Results show that post-tMCAO WBV significantly lessens cognitive deficits in rats of both sexes. Post-tMCAO WBV significantly decreased circulating pro-inflammatory cytokines and increased serum levels of irisin, a muscle-derived hormone that may play a role in brain metabolism and inflammation regulation, which suggests putative beneficial mechanisms of WBV.

5.
Transl Stroke Res ; 13(6): 898-912, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35306629

RESUMEN

Approximately 50% of stroke survivors experience gastrointestinal complications. The innate immune response plays a role in changes to the gut-brain axis after stroke. The purpose of this study is to examine the importance of inflammasome-mediated pyroptosis in disruption of the gut-brain axis after experimental stroke. B6129 mice were subjected to a closed-head photothrombotic stroke. We examined the time course of inflammasome protein expression in brain and intestinal lysate using western blot analysis at 1-, 3-, and 7-days post-injury for caspase-1, interleukin-1ß, nod-like receptor protein 3 (NLRP3), and apoptosis speck-like protein containing a caspase-recruiting domain (ASC) and gasdermin-D (GSDMD) cleavage. In a separate group of mice, we processed brain tissue 24 and 72 h after thrombotic stroke for immunohistochemical analysis of neuronal and endothelial cell pyroptosis. We examined intestinal tissue for morphological changes and pyroptosis of macrophages. We performed behavioral tests and assessed gut permeability changes to confirm functional changes after stroke. Our data show that thrombotic stroke induces inflammasome activation in the brain and intestinal tissue up to 7-day post-injury as well as pyroptosis of neurons, cerebral endothelial cells, and intestinal macrophages. We found that thrombotic stroke leads to neurocognitive and motor function deficits as well as increased gut permeability. Finally, the adoptive transfer of serum-derived EVs from stroke mice into naive induced inflammasome activation in intestinal tissues. Taken together, these results provide novel information regarding possible mechanisms underlying gut complications after stroke and the identification of new therapeutic targets for reducing the widespread consequences of ischemic brain injury.


Asunto(s)
Accidente Cerebrovascular , Accidente Cerebrovascular Trombótico , Animales , Ratones , Piroptosis , Inflamasomas/metabolismo , Células Endoteliales/metabolismo , Eje Cerebro-Intestino , Caspasas/metabolismo
6.
J Neurotrauma ; 38(5): 646-654, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32669032

RESUMEN

Traumatic brain injury (TBI) patients frequently develop cardiopulmonary system complications such as acute lung injury (ALI)/acute respiratory distress syndrome (ARDS). However, the mechanism by which TBI causes ALI/ARDS is not fully understood. Here, we used a severe TBI model to examine the effects of a low-molecular-weight heparin, enoxaparin, on inflammasome activation and lung injury damage. We investigated whether enoxaparin inhibits ALI and inflammasome signaling protein expression in the brain and lungs after TBI in mice. C57/BL6 mice were subjected to severe TBI and were treated with vehicle or 1 mg/kg of enoxaparin 30 min after injury. Lung and brain tissue were collected 24 h post-TBI and were analyzed by immunoblotting for expression of the inflammasome proteins, caspase-1 and interleukin (IL)-1ß. In addition, lung tissue was collected for histological analysis to determine ALI scoring and neutrophil and macrophage infiltration post-injury. Our data show that severe TBI induces increased expression of inflammasome proteins caspase-1 and IL-1ß in the brain and lungs of mice after injury. Treatment with enoxaparin attenuated inflammasome expression in the brain and lungs 24 h after injury. Enoxaparin significantly decreased ALI score as well as neutrophil and macrophage infiltration in lungs at 24 h after injury. This study demonstrates that enoxaparin attenuates ALI and inhibits inflammasome expression in the brain and lungs after TBI. These findings support the hypothesis that inhibition of the neural-respiratory inflammasome axis that is activated after TBI may have therapeutic potential.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/prevención & control , Anticoagulantes/uso terapéutico , Lesiones Traumáticas del Encéfalo/complicaciones , Enoxaparina/uso terapéutico , Inflamasomas/metabolismo , Lesión Pulmonar Aguda/etiología , Animales , Lesiones Traumáticas del Encéfalo/metabolismo , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila
7.
Sci Rep ; 11(1): 8791, 2021 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-33888735

RESUMEN

Hyperoxia-induced lung injury plays a key role in the development of bronchopulmonary dysplasia (BPD), characterized by inflammatory injury and impaired lung development in preterm infants. Although BPD is a predictor of poor neurodevelopmental outcomes, currently it is uncertain how lung injury contributes to brain injury in preterm infants. Extracellular vesicles (EVs) are a heterogeneous group of cell-derived membranous structures that regulate intercellular and inter-organ communications. Gasdermin D (GSDMD) has emerged as a key executor of inflammasome-mediated cell death and inflammation. In this study, we utilized a neonatal rat model of BPD to assess if hyperoxia stimulates lung release of circulating EVs and if these EVs induce lung and brain injury. We found that hyperoxia-exposed rats had elevated numbers of plasma-derived EVs compared to rats maintained in room air. These EVs also had increased cargos of surfactant protein C, a marker of type II alveolar epithelial cells (AEC), and the active (p30) form of GSDMD. When these EVs were adoptively transferred into normal newborn rats via intravenous injection, they were taken up both by lung and brain tissues. Moreover, EVs from hyperoxic animals induced not only the pathological hallmarks of BPD, but also brain inflammatory injury in recipient rats, as well as inducing cell death in cultured pulmonary vascular endothelial cells and neural stem cells (NSC). Similarly, hyperoxia-exposed cultured AEC-like cells released EVs that also contained increased GSDMD-p30 and these EVs induced pyroptotic cell death in NSC. Overall, these data indicate that hyperoxia-activated circulating EVs mediate a lung to brain crosstalk resulting in brain injury and suggest a mechanism that links lung injury and neurodevelopmental impairment in BPD infants.


Asunto(s)
Lesiones Encefálicas/metabolismo , Vesículas Extracelulares/metabolismo , Hiperoxia/metabolismo , Lesión Pulmonar/metabolismo , Animales , Animales Recién Nacidos , Femenino , Embarazo , Ratas , Ratas Sprague-Dawley
8.
Exp Neurol ; 323: 113080, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31626746

RESUMEN

Traumatic brain injury (TBI) is a leading cause of morbidity and mortality. Approximately 20-25% of TBI subjects develop Acute Lung Injury (ALI), but the pathomechanisms of TBI-induced ALI remain poorly defined. Currently, mechanical ventilation is the only therapeutic intervention for TBI-induced lung injury. Our recent studies have shown that the inflammasome plays an important role in the systemic inflammatory response leading to lung injury-post TBI. Here, we outline the role of the extracellular vesicle (EV)-mediated inflammasome signaling in the etiology of TBI-induced ALI. Furthermore, we evaluate the efficacy of a low molecular weight heparin (Enoxaparin, a blocker of EV uptake) and a monoclonal antibody against apoptosis speck-like staining protein containing a caspase recruitment domain (anti-ASC) as therapeutics for TBI-induced lung injury. We demonstate that activation of an EV-mediated Neural-Respiratory Inflammasome Axis plays an essential role in TBI-induced lung injury and disruption of this axis has therapeutic potential as a treatment strategy.


Asunto(s)
Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/inmunología , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/inmunología , Inflamasomas/inmunología , Lesión Pulmonar Aguda/fisiopatología , Animales , Lesiones Traumáticas del Encéfalo/fisiopatología , Vesículas Extracelulares/inmunología , Humanos , Inflamación/etiología , Inflamación/inmunología , Inflamación/fisiopatología
9.
CNS Neurosci Ther ; 25(12): 1308-1317, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31747126

RESUMEN

Ischemic stroke kills more women compared with men thus emphasizing a significant sexual dimorphism in ischemic pathophysiological outcomes. However, the mechanisms behind this sexual dimorphism are yet to be fully understood. It is well established that cerebral ischemia activates a variety of inflammatory cascades and that microglia are the primary immune cells of the brain. After ischemic injury, microglia are activated and play a crucial role in progression and resolution of the neuroinflammatory response. In recent years, research has focused on the role that microglia play in this sexual dimorphism that exists in the response to central nervous system (CNS) injury. Evidence suggests that the molecular mechanisms leading to microglial activation and polarization of phenotypes may be influenced by sex, therefore causing a difference in the pro/anti-inflammatory responses after CNS injury. Here, we review advances highlighting that sex differences in microglia are an important factor in the inflammatory responses that are seen after ischemic injury. We discuss the main differences between microglia in the healthy and diseased developing, adult, and aging brain. We also focus on the dimorphism that exists between males and females in microglial-induced inflammation and energy metabolism after CNS injury. Finally, we describe how all of the current research and literature regarding sex differences in microglia contribute to the differences in poststroke responses between males and females.


Asunto(s)
Isquemia Encefálica/patología , Microglía/patología , Caracteres Sexuales , Accidente Cerebrovascular/patología , Animales , Femenino , Humanos , Masculino
10.
Cells ; 8(1)2019 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-30669285

RESUMEN

Approximately 30% of traumatic brain injured patients suffer from acute lung injury or acute respiratory distress syndrome. Our previous work revealed that extracellular vesicle (EV)-mediated inflammasome signaling plays a crucial role in the pathophysiology of traumatic brain injury (TBI)-induced lung injury. Here, serum-derived EVs from severe TBI patients were analyzed for particle size, concentration, origin, and levels of the inflammasome component, an apoptosis-associated speck-like protein containing a caspase-recruiting domain (ASC). Serum ASC levels were analyzed from EV obtained from patients that presented lung injury after TBI and compared them to EV obtained from patients that did not show any signs of lung injury. EVs were co-cultured with lung human microvascular endothelial cells (HMVEC-L) to evaluate inflammasome activation and endothelial cell pyroptosis. TBI patients had a significant increase in the number of serum-derived EVs and levels of ASC. Severe TBI patients with lung injury had a significantly higher level of ASC in serum and serum-derived EVs compared to individuals without lung injury. Only EVs isolated from head trauma patients with gunshot wounds were of neural origin. Delivery of serum-derived EVs to HMVEC-L activated the inflammasome and resulted in endothelial cell pyroptosis. Thus, serum-derived EVs and inflammasome proteins play a critical role in the pathogenesis of TBI-induced lung injury, supporting activation of an EV-mediated neural-respiratory inflammasome axis in TBI-induced lung injury.


Asunto(s)
Lesiones Traumáticas del Encéfalo/complicaciones , Pulmón/patología , Piroptosis , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/metabolismo , Lesiones Traumáticas del Encéfalo/sangre , Proteínas Adaptadoras de Señalización CARD/metabolismo , Células Endoteliales/metabolismo , Vesículas Extracelulares/metabolismo , Femenino , Humanos , Inflamasomas/metabolismo , Pulmón/irrigación sanguínea , Lesión Pulmonar/etiología , Masculino , Persona de Mediana Edad , Curva ROC , Adulto Joven
12.
Front Mol Neurosci ; 11: 309, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30233311

RESUMEN

The inflammasome is a key contributor to the inflammatory innate immune response after stroke. We have previously shown that inflammasome proteins are released in extracellular vesicles (EV) after brain and spinal cord injury. In addition, we have shown that inflammasome proteins offer great promise as biomarkers of central nervous system (CNS) injury following brain trauma. In the present study, we used a Simple Plex Assay (Protein Simple), a novel multi-analyte automated microfluidic immunoassay platform, to analyze serum and serum-derived EV samples from stroke patients and control subjects for inflammasome protein levels of caspase-1, apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC), Interleukins (IL)-1ß, and (IL)-18. Receiver operator characteristic (ROC) curves with associated confidence intervals obtained from the analysis of serum samples revealed that the area under the curve (AUC) for ASC was 0.99 with a confidence interval between 0.9914 and 1.004, whereas the AUC for caspase-1, IL-1ß, and IL-18 were 0.75, 0.61, and 0.67, respectively. Thus, these data indicate that ASC is a potential biomarker of stroke and highlight the role of the inflammasome in the inflammatory response after brain ischemia.

13.
PLoS One ; 13(12): e0210128, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30596792

RESUMEN

BACKGROUND: The inflammasome plays an important role in the inflammatory innate immune response after central nervous system (CNS) injury. Inhibition of the inflammasome after traumatic brain injury (TBI) results in improved outcomes by lowering the levels of caspase-1 and interleukin (IL)-1b. We have previously shown that inflammasome proteins are elevated in the cerebrospinal fluid (CSF) of patients with TBI and that higher levels of these proteins were consistent with poorer outcomes after TBI when compared to patients that presented these inflammasome proteins at lower levels. METHODS AND FINDINGS: Here we extend our work by analyzing serum from 21 TBI patients and CSF from 18 TBI patients compared to 120 serum samples and 30 CSF samples from no-TBI donor controls for the expression of caspase-1, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), interleukin(IL)-1b and IL-18. Analysis was carried out using the Ella Simple Plex system (Protein Simple) to determine the sensitivity and specificity of inflammasome proteins as biomarkers of TBI. Receiver operator characteristic (ROC) curves, confidence intervals and likelihood ratios for each biomarker was determined. ROC curves, confidence intervals, sensitivity and specificity for each biomarker examined revealed that caspase-1 (0.93 area under the curve (AUC)) and ASC (0.90 AUC) in serum and ASC (1.0 AUC) and IL-18 (0.84 AUC) in CSF are promising biomarkers of TBI pathology. Importantly, higher protein levels (above 547.6 pg/ml) of ASC (0.91 AUC) were consistent with poorer outcomes after TBI as determined by the Glasgow Outcome Scale-Extended (GOSE). CONCLUSION: These findings indicate that inflammasome proteins are excellent diagnostic and predictive biomarkers of TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Proteínas Adaptadoras de Señalización CARD , Caspasa 1 , Inflamasomas , Interleucina-18 , Interleucina-1beta , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Biomarcadores/líquido cefalorraquídeo , Lesiones Traumáticas del Encéfalo/sangre , Lesiones Traumáticas del Encéfalo/líquido cefalorraquídeo , Proteínas Adaptadoras de Señalización CARD/sangre , Proteínas Adaptadoras de Señalización CARD/líquido cefalorraquídeo , Caspasa 1/sangre , Caspasa 1/líquido cefalorraquídeo , Femenino , Humanos , Inflamasomas/sangre , Inflamasomas/líquido cefalorraquídeo , Interleucina-18/sangre , Interleucina-18/líquido cefalorraquídeo , Interleucina-1beta/sangre , Interleucina-1beta/líquido cefalorraquídeo , Masculino , Persona de Mediana Edad
14.
J Neurotrauma ; 35(17): 2067-2076, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29648974

RESUMEN

Approximately 20-25% of traumatic brain injury (TBI) subjects develop acute lung injury (ALI), but the pathomechanisms of TBI-induced ALI remain poorly defined. Our previous work has shown that the inflammasome plays a critical role in TBI-induced secondary pathophysiology and that inflammasome proteins are released in extracellular vesicles (EV) after TBI. Here we investigated whether EV-mediated inflammasome signaling contributed to the etiology of TBI-induced ALI. C57/BL6 male mice were subjected to controlled cortical impact (CCI), and the brains and lungs were examined for inflammasome activation and ALI at 4 and 24 h after TBI. We show that TBI releases EV containing inflammasome proteins into serum that target the lung to cause ALI, supporting activation of a neural-respiratory-inflammasome axis. Administration of a low-molecular-weight heparin (enoxaparin, a blocker of EV uptake) or treatment with a monoclonal antibody against apoptosis speck-like staining protein containing a caspase recruitment domain (anti-ASC) after adoptive transfer of EV isolated from TBI-injured mice significantly inhibited inflammasome activation in the lungs of recipient mice resulting in improved ALI scores.This axis constitutes an important arm of the innate inflammatory response in lung pathology after TBI and targeting this axis represents a novel therapeutic treatment for TBI-induced ALI.


Asunto(s)
Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/fisiopatología , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/fisiopatología , Inflamasomas , Sistema Nervioso/fisiopatología , Sistema Respiratorio/fisiopatología , Animales , Anticoagulantes/uso terapéutico , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Enoxaparina/uso terapéutico , Espacio Extracelular/metabolismo , Proteína HMGB1/biosíntesis , Proteína HMGB1/genética , Inmunidad Innata , Masculino , Ratones , Ratones Endogámicos C57BL
15.
Neonatology ; 111(3): 280-288, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28013306

RESUMEN

BACKGROUND: Inflammatory injury, particularly the production of active interleukin (IL)-1ß plays a major role in the pathogenesis of bronchopulmonary dysplasia (BPD) in preterm infants. The release of active IL-1ß is controlled by posttranscriptional modifications of its proform (pro-IL-1ß) through the inflammasome. Rac1 is a member of the Rho family of GTPases that regulate the inflammatory process. OBJECTIVE: This study tested the hypothesis that Rac1 signaling increases inflammasome activation that results in damaging inflammation, and that the inhibition of Rac1 signaling prevents lung injury, by inhibiting inflammasome activation in a newborn rat model of BPD induced by hyperoxia. METHODS: Newborn rat pups were exposed to room air or hyperoxia (85% O2) and received daily intraperitoneal injections of placebo (normal saline) or NSC23766, a specific Rac1 inhibitor, for 10 days. The effects on lung inflammation, alveolarization, vascular development, vascular remodeling, right ventricular systolic pressure, and right ventricular hypertrophy (RVH) were then assessed. RESULTS: Hyperoxia exposure upregulated Rac1 and increased the production of active IL-1ß, which was accompanied by increasing expression of the inflammasome. In addition, hyperoxia induced the pathological hallmarks of BPD. However, treatment with NSC23766 significantly decreased inflammasome activation and macrophage infiltration, improved alveolar and vascular development, and reduced pulmonary vascular remodeling and RVH. CONCLUSION: These results indicate that Rac1 signaling regulates the expression of the inflammasome and plays a pivotal role in the pathogenesis of hyperoxia-induced neonatal lung injury. Therefore, targeting Rac1 signaling may provide a novel strategy to prevent and treat BPD in preterm infants.


Asunto(s)
Hiperoxia/complicaciones , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Pulmón/patología , Proteína de Unión al GTP rac1/metabolismo , Aminoquinolinas/farmacología , Animales , Animales Recién Nacidos , Displasia Broncopulmonar/tratamiento farmacológico , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Hipertensión Pulmonar/tratamiento farmacológico , Hipertrofia Ventricular Derecha/tratamiento farmacológico , Inflamasomas/efectos de los fármacos , Interleucina-1beta/efectos de los fármacos , Pirimidinas/farmacología , Ratas , Proteína de Unión al GTP rac1/antagonistas & inhibidores
16.
J Neurotrauma ; 33(16): 1522-34, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-26541248

RESUMEN

Reactions of both astrocytes and microglia to central nervous system injury can be beneficial or detrimental to recovery. To gain insights into the functional importance of gliosis, we developed a new model of adolescent closed-head injury (CHI) and interrogated the behavioral, physiological, and cellular outcomes after a concussive CHI in leukemia inhibitory factor (LIF) haplodeficient mice. These mice were chosen because LIF is important for astrocyte and microglial activation. Behaviorally, the LIF haplodeficient animals were equally impaired 4 h after the injury, but in the subsequent 2 weeks, the LIF haplodeficient mice acquired more severe motor and sensory deficits, compared with wild type mice. The prolonged accumulation of neurological impairment was accompanied by desynchronization of the gliotic response, increased cell death, axonal degeneration, diminished callosal compound action potential, and hypomyelination. Our results clearly show that LIF is an essential injury-induced cytokine that is required to prevent the propagation of secondary neurodegeneration.


Asunto(s)
Citocinas/fisiología , Traumatismos Cerrados de la Cabeza/metabolismo , Traumatismos Cerrados de la Cabeza/patología , Traumatismos Cerrados de la Cabeza/fisiopatología , Factor Inhibidor de Leucemia/fisiología , Animales , Citocinas/deficiencia , Modelos Animales de Enfermedad , Femenino , Factor Inhibidor de Leucemia/deficiencia , Masculino , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA