Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2023 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-37905154

RESUMO

Microglia and astrocytes play an important role in the neuroinflammatory response and contribute to both the destruction of neighboring tissue as well as the resolution of inflammation following stroke. These reactive glial cells are highly heterogeneous at both the transcriptomic and functional level. Depending upon the stimulus, microglia and astrocytes mount a complex, and specific response composed of distinct microglial and astrocyte substates. These substates ultimately drive the landscape of the initiation and recovery from the adverse stimulus. In one state, inflammation- and damage-induced microglia release tumor necrosis factor (TNF), interleukin 1α (IL1α), and complement component 1q (C1q), together 'TIC'. This cocktail of cytokines drives astrocytes into a neurotoxic reactive astrocyte (nRA) substate. This nRA substate is associated with loss of many physiological astrocyte functions (e.g., synapse formation and maturation, phagocytosis, among others), as well as a gain-of-function release of neurotoxic long-chain fatty acids which kill neighboring cells. Here we report that transgenic removal of TIC led to reduction of gliosis, infarct expansion, and worsened functional deficits in the acute and delayed stages following stroke. Our results suggest that TIC cytokines, and likely nRAs play an important role that may maintain neuroinflammation and inhibit functional motor recovery after ischemic stroke. This is the first report that this paradigm is relevant in stroke and that therapies against nRAs may be a novel means to treat patients. Since nRAs are evolutionarily conserved from rodents to humans and present in multiple neurodegenerative diseases and injuries, further identification of mechanistic role of nRAs will lead to a better understanding of the neuroinflammatory response and the development of new therapies.

2.
Front Mol Neurosci ; 16: 1305949, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38240014

RESUMO

Microglia and astrocytes play an important role in the neuroinflammatory response and contribute to both the destruction of neighboring tissue as well as the resolution of inflammation following stroke. These reactive glial cells are highly heterogeneous at both the transcriptomic and functional level. Depending upon the stimulus, microglia and astrocytes mount a complex, and specific response composed of distinct microglial and astrocyte substates. These substates ultimately drive the landscape of the initiation and recovery from the adverse stimulus. In one state, inflammation- and damage-induced microglia release tumor necrosis factor (TNF), interleukin 1α (IL1α), and complement component 1q (C1q), together "TIC." This cocktail of cytokines drives astrocytes into a neurotoxic reactive astrocyte (nRA) substate. This nRA substate is associated with loss of many physiological astrocyte functions (e.g., synapse formation and maturation, phagocytosis, among others), as well as a gain-of-function release of neurotoxic long-chain fatty acids which kill neighboring cells. Here we report that transgenic removal of TIC led to reduction of gliosis, infarct expansion, and worsened functional deficits in the acute and delayed stages following stroke. Our results suggest that TIC cytokines, and likely nRAs play an important role that may maintain neuroinflammation and inhibit functional motor recovery after ischemic stroke. This is the first report that this paradigm is relevant in stroke and that therapies against nRAs may be a novel means to treat patients. Since nRAs are evolutionarily conserved from rodents to humans and present in multiple neurodegenerative diseases and injuries, further identification of mechanistic role of nRAs will lead to a better understanding of the neuroinflammatory response and the development of new therapies.

3.
J Neuroinflammation ; 18(1): 199, 2021 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-34511110

RESUMO

BACKGROUND: Patients with chronic inflammatory disorders such as inflammatory bowel disease frequently experience neurological complications including epilepsy, depression, attention deficit disorders, migraines, and dementia. However, the mechanistic basis for these associations is unknown. Given that many patients are unresponsive to existing medications or experience debilitating side effects, novel therapeutics that target the underlying pathophysiology of these conditions are urgently needed. METHODS: Because intestinal disorders such as inflammatory bowel disease are robustly associated with neurological symptoms, we used three different mouse models of colitis to investigate the impact of peripheral inflammatory disease on the brain. We assessed neuronal hyperexcitability, which is associated with many neurological symptoms, by measuring seizure threshold in healthy and colitic mice. We profiled the neuroinflammatory phenotype of colitic mice and used depletion and neutralization assays to identify the specific mediators responsible for colitis-induced neuronal hyperexcitability. To determine whether our findings in murine models overlapped with a human phenotype, we performed gene expression profiling, pathway analysis, and deconvolution on microarray data from hyperexcitable human brain tissue from patients with epilepsy. RESULTS: We observed that murine colitis induces neuroinflammation characterized by increased pro-inflammatory cytokine production, decreased tight junction protein expression, and infiltration of monocytes and neutrophils into the brain. We also observed sustained neuronal hyperexcitability in colitic mice. Colitis-induced neuronal hyperexcitability was ameliorated by neutrophil depletion or TNFα blockade. Gene expression profiling of hyperexcitable brain tissue resected from patients with epilepsy also revealed a remarkably similar pathology to that seen in the brains of colitic mice, including neutrophil infiltration and high TNFα expression. CONCLUSIONS: Our results reveal neutrophils and TNFα as central regulators of neuronal hyperexcitability of diverse etiology. Thus, there is a strong rationale for evaluating anti-inflammatory agents, including clinically approved TNFα inhibitors, for the treatment of neurological and psychiatric symptoms present in, and potentially independent of, a diagnosed inflammatory disorder.


Assuntos
Colite , Epilepsia , Animais , Encéfalo/metabolismo , Colite/induzido quimicamente , Modelos Animais de Doenças , Epilepsia/complicações , Humanos , Camundongos , Neurônios , Neutrófilos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
4.
J Nucl Med ; 60(1): 122-128, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29976695

RESUMO

Neuroinflammation plays a key role in neuronal injury after ischemic stroke. PET imaging of translocator protein 18 kDa (TSPO) permits longitudinal, noninvasive visualization of neuroinflammation in both preclinical and clinical settings. Many TSPO tracers have been developed, however, it is unclear which tracer is the most sensitive and accurate for monitoring the in vivo spatiotemporal dynamics of neuroinflammation across applications. Hence, there is a need for head-to-head comparisons of promising TSPO PET tracers across different disease states. Accordingly, the aim of this study was to directly compare 2 promising second-generation TSPO tracers, 11C-DPA-713 and 18F-GE-180, for the first time at acute and chronic time points after ischemic stroke. Methods: After distal middle cerebral artery occlusion or sham surgery, mice underwent consecutive PET/CT imaging with 11C-DPA-713 and 18F-GE-180 at 2, 6, and 28 d after stroke. T2-weighted MR images were acquired to enable delineation of ipsilateral (infarct) and contralateral brain regions of interest (ROIs). PET/CT images were analyzed by calculating percentage injected dose per gram in MR-guided ROIs. SUV ratios were determined using the contralateral thalamus (SUVTh) as a pseudoreference region. Ex vivo autoradiography and immunohistochemistry were performed to verify in vivo findings. Results: Significantly increased tracer uptake was observed in the ipsilateral compared with contralateral ROI (SUVTh, 50-60 min summed data) at acute and chronic time points using 11C-DPA-713 and 18F-GE-180. Ex vivo autoradiography confirmed in vivo findings demonstrating increased TSPO tracer uptake in infarcted versus contralateral brain tissue. Importantly, a significant correlation was identified between microglial/macrophage activation (cluster of differentiation 68 immunostaining) and 11C-DPA-713- PET signal, which was not evident with 18F-GE-180. No significant correlations were observed between TSPO PET and activated astrocytes (glial fibrillary acidic protein immunostaining). Conclusion:11C-DPA-713 and 18F-GE-180 PET enable detection of neuroinflammation at acute and chronic time points after cerebral ischemia in mice. 11C-DPA-713 PET reflects the extent of microglial activation in infarcted distal middle cerebral artery occlusion mouse brain tissue more accurately than 18F-GE-180 and appears to be slightly more sensitive. These results highlight the potential of 11C-DPA-713 for tracking microglial activation in vivo after stroke and warrant further investigation in both preclinical and clinical settings.


Assuntos
Acetamidas , Isquemia Encefálica/complicações , Carbazóis , Tomografia por Emissão de Pósitrons/métodos , Pirazóis , Pirimidinas , Receptores de GABA/metabolismo , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/diagnóstico por imagem , Doença Aguda , Animais , Radioisótopos de Carbono , Doença Crônica , Modelos Animais de Doenças , Radioisótopos de Flúor , Inflamação/diagnóstico por imagem , Camundongos , Traçadores Radioativos , Acidente Vascular Cerebral/metabolismo
5.
Nature ; 541(7638): 481-487, 2017 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-28099414

RESUMO

Reactive astrocytes are strongly induced by central nervous system (CNS) injury and disease, but their role is poorly understood. Here we show that a subtype of reactive astrocytes, which we termed A1, is induced by classically activated neuroinflammatory microglia. We show that activated microglia induce A1 astrocytes by secreting Il-1α, TNF and C1q, and that these cytokines together are necessary and sufficient to induce A1 astrocytes. A1 astrocytes lose the ability to promote neuronal survival, outgrowth, synaptogenesis and phagocytosis, and induce the death of neurons and oligodendrocytes. Death of axotomized CNS neurons in vivo is prevented when the formation of A1 astrocytes is blocked. Finally, we show that A1 astrocytes are abundant in various human neurodegenerative diseases including Alzheimer's, Huntington's and Parkinson's disease, amyotrophic lateral sclerosis and multiple sclerosis. Taken together these findings help to explain why CNS neurons die after axotomy, strongly suggest that A1 astrocytes contribute to the death of neurons and oligodendrocytes in neurodegenerative disorders, and provide opportunities for the development of new treatments for these diseases.


Assuntos
Astrócitos/classificação , Astrócitos/patologia , Morte Celular , Sistema Nervoso Central/patologia , Microglia/patologia , Neurônios/patologia , Animais , Astrócitos/metabolismo , Axotomia , Técnicas de Cultura de Células , Sobrevivência Celular , Complemento C1q/metabolismo , Progressão da Doença , Humanos , Inflamação/patologia , Interleucina-1alfa/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Doenças Neurodegenerativas/patologia , Oligodendroglia/patologia , Fagocitose , Fenótipo , Ratos , Ratos Sprague-Dawley , Sinapses/patologia , Toxinas Biológicas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
6.
J Neurosci ; 35(5): 2133-45, 2015 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-25653369

RESUMO

Each year, 10 million people worldwide survive the neurologic injury associated with a stroke. Importantly, stroke survivors have more than twice the risk of subsequently developing dementia compared with people who have never had a stroke. The link between stroke and the later development of dementia is not understood. There are reports of oligoclonal bands in the CSF of stroke patients, suggesting that in some people a B-lymphocyte response to stroke may occur in the CNS. Therefore, we tested the hypothesis that a B-lymphocyte response to stroke could contribute to the onset of dementia. We discovered that, in mouse models, activated B-lymphocytes infiltrate infarcted tissue in the weeks after stroke. B-lymphocytes undergo isotype switching, and IgM, IgG, and IgA antibodies are found in the neuropil adjacent to the lesion. Concurrently, mice develop delayed deficits in LTP and cognition. Genetic deficiency, and the pharmacologic ablation of B-lymphocytes using an anti-CD20 antibody, prevents the appearance of delayed cognitive deficits. Furthermore, immunostaining of human postmortem tissue revealed that a B-lymphocyte response to stroke also occurs in the brain of some people with stroke and dementia. These data suggest that some stroke patients may develop a B-lymphocyte response to stroke that contributes to dementia, and is potentially treatable with FDA-approved drugs that target B cells.


Assuntos
Subpopulações de Linfócitos B/imunologia , Demência/etiologia , Infarto da Artéria Cerebral Média/imunologia , Idoso , Animais , Estudos de Casos e Controles , Demência/imunologia , Demência/fisiopatologia , Feminino , Humanos , Imunoglobulinas/imunologia , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/fisiopatologia , Potenciação de Longa Duração , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
7.
Neurosci Lett ; 584: 236-40, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25449870

RESUMO

Ferumoxytol is an ultrasmall superparamagnetic iron oxide (USPIO) nanoparticle that is FDA-approved as an intravenous iron replacement therapy for the treatment of iron deficiency anemia in patients with chronic kidney disease. Ferumoxytol has also been used as a contrast agent for cerebral blood volume mapping by magnetic resonance imaging (MRI), which suggests it could be used for imaging hemodynamic abnormalities after stroke. However, circulating macrophages can internalize USPIOs, and recent data indicate that the accumulation of iron in macrophages can lead them to adopt the M1 pro-inflammatory phenotype. Therefore, the uptake of intravenously administered iron particles by circulating macrophages that home to the stroke core could potentially alter the inflammatory response to stroke. To test this possibility in vivo we administered a dose of ferumoxytol previously used to obtain cerebral blood volume maps in healthy humans by steady-state susceptibility contrast (SSC) MRI to BALB/cJ mice 48h after stroke and examined cytokine levels, microglial/macrophage activation, and lesion volume in the brain 5 days later. Treatment with ferumoxytol did not lead to any differences in these parameters. These data indicate that the use of ferumoxytol as a contrast agent for brain imaging after stroke does not alter the inflammatory response to stroke in mice, and is therefore unlikely to do so in human subjects.


Assuntos
Infarto Encefálico/patologia , Meios de Contraste/toxicidade , Óxido Ferroso-Férrico/toxicidade , Acidente Vascular Cerebral/patologia , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Infarto Encefálico/imunologia , Infarto Encefálico/metabolismo , Citocinas/metabolismo , Inflamação/induzido quimicamente , Inflamação/metabolismo , Ferro/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Acidente Vascular Cerebral/imunologia , Acidente Vascular Cerebral/metabolismo
8.
Glia ; 62(8): 1227-40, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24733756

RESUMO

Astrocytes limit inflammation after CNS injury, at least partially by physically containing it within an astrocytic scar at the injury border. We report here that astrocytic transforming growth factor-beta (TGFß) signaling is a second, distinct mechanism that astrocytes utilize to limit neuroinflammation. TGFßs are anti-inflammatory and neuroprotective cytokines that are upregulated subacutely after stroke, during a clinically accessible time window. We have previously demonstrated that TGFßs signal to astrocytes, neurons and microglia in the stroke border days after stroke. To investigate whether TGFß affects astrocyte immunoregulatory functions, we engineered "Ast-Tbr2DN" mice where TGFß signaling is inhibited specifically in astrocytes. Despite having a similar infarct size to wildtype controls, Ast-Tbr2DN mice exhibited significantly more neuroinflammation during the subacute period after distal middle cerebral occlusion (dMCAO) stroke. The peri-infarct cortex of Ast-Tbr2DN mice contained over 60% more activated CD11b(+) monocytic cells and twice as much immunostaining for the activated microglia and macrophage marker CD68 than controls. Astrocytic scarring was not altered in Ast-Tbr2DN mice. However, Ast-Tbr2DN mice were unable to upregulate TGF-ß1 and its activator thrombospondin-1 2 days after dMCAO. As a result, the normal upregulation of peri-infarct TGFß signaling was blunted in Ast-Tbr2DN mice. In this setting of lower TGFß signaling and excessive neuroinflammation, we observed worse motor outcomes and late infarct expansion after photothrombotic motor cortex stroke. Taken together, these data demonstrate that TGFß signaling is a molecular mechanism by which astrocytes limit neuroinflammation, activate TGFß in the peri-infarct cortex and preserve brain function during the subacute period after stroke.


Assuntos
Astrócitos/imunologia , Córtex Cerebral/imunologia , Infarto da Artéria Cerebral Média/imunologia , Acidente Vascular Cerebral/imunologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Astrócitos/patologia , Antígeno CD11b/metabolismo , Córtex Cerebral/patologia , Modelos Animais de Doenças , Feminino , Infarto da Artéria Cerebral Média/patologia , Macrófagos/patologia , Macrófagos/fisiologia , Camundongos Transgênicos , Microglia/imunologia , Microglia/patologia , Monócitos/patologia , Monócitos/fisiologia , Atividade Motora/fisiologia , Neuroimunomodulação/fisiologia , Transdução de Sinais , Acidente Vascular Cerebral/patologia , Trombospondina 1/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta1/metabolismo
9.
Methods Mol Biol ; 1135: 103-10, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24510858

RESUMO

Here we provide a standardized protocol for performing distal middle cerebral artery occlusion (DMCAO) in mice. DMCAO is a method of inducing permanent focal ischemia that is commonly used as a rodent stroke model. To perform DMCAO a temporal craniotomy is performed, and the middle cerebral artery (MCA) is permanently ligated at a point downstream of the lenticulostriate branches. The size of the lesion produced by this surgery is strain dependent. In C57BL/6J mice, DMCAO produces an infarct predominantly restricted to the barrel region of the somatosensory cortex, but in BALB/cJ mice, DMCAO generates a much larger lesion that incorporates more of the somatosensory cortex and part of the M1 region of the motor cortex. The larger lesion produced by DMCAO in BALB/cJ mice produces a clearer sensorimotor deficit, which is useful for investigating recovery from stroke. We also describe how to modify DMCAO in C57BL/6J mice with the application of hypoxia to generate a lesion and sensorimotor deficit that are similar in size to those produced by DMCAO alone in BALB/cJ mice. This is extremely useful for stroke experiments that require a robust sensorimotor deficit in transgenic mice created on a C57BL/6J background.


Assuntos
Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/patologia , Animais , Córtex Cerebral/irrigação sanguínea , Infarto da Artéria Cerebral Média/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Artéria Cerebral Média/patologia
11.
J Neuroinflammation ; 7: 62, 2010 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-20937129

RESUMO

BACKGROUND: TGFß is both neuroprotective and a key immune system modulator and is likely to be an important target for future stroke therapy. The precise function of increased TGF-ß1 after stroke is unknown and its pleiotropic nature means that it may convey a neuroprotective signal, orchestrate glial scarring or function as an important immune system regulator. We therefore investigated the time course and cell-specificity of TGFß signaling after stroke, and whether its signaling pattern is altered by gender and aging. METHODS: We performed distal middle cerebral artery occlusion strokes on 5 and 18 month old TGFß reporter mice to get a readout of TGFß responses after stroke in real time. To determine which cell type is the source of increased TGFß production after stroke, brain sections were stained with an anti-TGFß antibody, colocalized with markers for reactive astrocytes, neurons, and activated microglia. To determine which cells are responding to TGFß after stroke, brain sections were double-labelled with anti-pSmad2, a marker of TGFß signaling, and markers of neurons, oligodendrocytes, endothelial cells, astrocytes and microglia. RESULTS: TGFß signaling increased 2 fold after stroke, beginning on day 1 and peaking on day 7. This pattern of increase was preserved in old animals and absolute TGFß signaling in the brain increased with age. Activated microglia and macrophages were the predominant source of increased TGFß after stroke and astrocytes and activated microglia and macrophages demonstrated dramatic upregulation of TGFß signaling after stroke. TGFß signaling in neurons and oligodendrocytes did not undergo marked changes. CONCLUSIONS: We found that TGFß signaling increases with age and that astrocytes and activated microglia and macrophages are the main cell types that undergo increased TGFß signaling in response to post-stroke increases in TGFß. Therefore increased TGFß after stroke likely regulates glial scar formation and the immune response to stroke.


Assuntos
Envelhecimento/metabolismo , Astrócitos/metabolismo , Encéfalo/metabolismo , Macrófagos/metabolismo , Acidente Vascular Cerebral/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Envelhecimento/imunologia , Análise de Variância , Animais , Astrócitos/imunologia , Western Blotting , Encéfalo/imunologia , Feminino , Imunofluorescência , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Transgênicos , Microglia/imunologia , Microglia/metabolismo , Neurônios/imunologia , Neurônios/metabolismo , Transdução de Sinais , Acidente Vascular Cerebral/imunologia , Fator de Crescimento Transformador beta/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA