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










Base de datos
Intervalo de año de publicación
1.
Mol Ther ; 32(3): 783-799, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38196192

RESUMEN

We recently described a novel ribosome-based regulatory mechanism/checkpoint that controls innate immune gene translation and microglial activation in non-sterile inflammation orchestrated by RNA binding protein SRSF3. Here we describe a role of SRSF3 in the regulation of microglia/macrophage activation phenotypes after experimental stroke. Using a model-system for analysis of the dynamic translational state of microglial ribosomes we show that 24 h after stroke highly upregulated immune mRNAs are not translated resulting in a marked dissociation of mRNA and protein networks in activated microglia/macrophages. Next, microglial activation after stroke was characterized by a robust increase in pSRSF3/SRSF3 expression levels. Targeted knockdown of SRSF3 using intranasal delivery of siRNA 24 h after stroke caused a marked knockdown of endogenous protein. Further analyses revealed that treatment with SRSF3-siRNA alleviated translational arrest of selected genes and induced a transient but significant increase in innate immune signaling and IBA1+ immunoreactivity peaking 5 days after initial injury. Importantly, delayed SRSF3-mediated increase in immune signaling markedly reduced the size of ischemic lesion measured 7 days after stroke. Together, our findings suggest that targeting SRSF3 and immune mRNA translation may open new avenues for molecular/therapeutic reprogramming of innate immune response after ischemic injury.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular , Humanos , Microglía/metabolismo , Isquemia Encefálica/genética , Isquemia Encefálica/terapia , Macrófagos/metabolismo , Accidente Cerebrovascular/patología , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Factores de Empalme Serina-Arginina/genética , Factores de Empalme Serina-Arginina/metabolismo
2.
Metabolites ; 13(11)2023 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-37999253

RESUMEN

Aging is not a disease; it is a natural evolution of human physiology. Medical advances have extended our life expectancy, but chronic diseases and geriatric syndrome continue to affect the increasingly aging population. Yet modern medicine perpetuates an approach based on treatment rather than prevention and education. In order to help solve this ever-growing problem, a new discipline has emerged: lifestyle medicine. Nutrition, physical activity, stress management, restorative sleep, social connection, and avoidance of risky substances are the pillars on which lifestyle medicine is founded. The aim of this discipline is to increase healthspan and reduce the duration of morbidity by making changes to our lifestyle. In this review, we propose the use of klotho protein as a novel biomarker for lifestyle medicine in order to quantify and monitor the health status of individuals, as no integrative tool currently exists.

3.
Int J Mol Sci ; 24(6)2023 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-36982140

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a clinically highly heterogeneous disease with a survival rate ranging from months to decades. Evidence suggests that a systemic deregulation of immune response may play a role and affect disease progression. Here, we measured 62 different immune/metabolic mediators in plasma of sporadic ALS (sALS) patients. We show that, at the protein level, the majority of immune mediators including a metabolic sensor, leptin, were significantly decreased in the plasma of sALS patients and in two animal models of the disease. Next, we found that a subset of patients with rapidly progressing ALS develop a distinct plasma assess immune-metabolic molecular signature characterized by a differential increase in soluble tumor necrosis factor receptor II (sTNF-RII) and chemokine (C-C motif) ligand 16 (CCL16) and further decrease in the levels of leptin, mostly dysregulated in male patients. Consistent with in vivo findings, exposure of human adipocytes to sALS plasma and/or sTNF-RII alone, induced a significant deregulation in leptin production/homeostasis and was associated with a robust increase in AMP-activated protein kinase (AMPK) phosphorylation. Conversely, treatment with an AMPK inhibitor restored leptin production in human adipocytes. Together, this study provides evidence of a distinct plasma immune profile in sALS which affects adipocyte function and leptin signaling. Furthermore, our results suggest that targeting the sTNF-RII/AMPK/leptin pathway in adipocytes may help restore assess immune-metabolic homeostasis in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , Leptina , Animales , Humanos , Masculino , Proteínas Quinasas Activadas por AMP , Receptores del Factor de Necrosis Tumoral , Homeostasis
4.
Mol Neurodegener ; 16(1): 1, 2021 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33413517

RESUMEN

BACKGROUND: TDP-43 proteinopathy is a pathological hallmark of many neurodegenerative diseases including amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). So far, there is no therapy available for these neurodegenerative diseases. In addition, the impact of TDP-43 proteinopathy on neuronal translational profile also remains unknown. METHODS: Biochemical, immunohistology and assay-based studies were done with cell cultures and transgenic mice models. We also used Ribotag with microarray and proteomic analysis to determine the neuronal translational profile in the mice model of ALS/FTD. RESULTS: Here, we report that oral administration of a novel analog (IMS-088) of withaferin-A, an antagonist of nuclear factor kappa-B (NF-ĸB) essential modulator (NEMO), induced autophagy and reduced TDP-43 proteinopathy in the brain and spinal cord of transgenic mice expressing human TDP-43 mutants, models of ALS/FTD. Treatment with IMS-088 ameliorated cognitive impairment, reduced gliosis in the brain of ALS/FTD mouse models. With the Ribotrap method, we investigated the impact of TDP-43 proteinopathy and IMS-088 treatment on the translation profile of neurons of one-year old hTDP-43A315T mice. TDP-43 proteinopathy caused translational dysregulation of specific mRNAs including translational suppression of neurofilament mRNAs resulting in 3 to 4-fold decrease in levels type IV neurofilament proteins. Oral administration of IMS-088 rescued the translational defects associated with TDP-43 proteinopathy and restored the synthesis of neurofilament proteins, which are essential for axon integrity and synaptic function. CONCLUSIONS: Our study revealed that induction of autophagy reduces TDP-43 pathology and ameliorates the translational defect seen in mice models of ALS/FTD. Based on these results, we suggest IMS-088 and perhaps other inducers of autophagy should be considered as potential therapeutics for neurodegenerative disorders with TDP-43 proteinopathies.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Autofagia/fisiología , Demencia Frontotemporal/metabolismo , ARN Mensajero/metabolismo , Proteinopatías TDP-43/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Demencia Frontotemporal/patología , Humanos , Filamentos Intermedios/metabolismo , Filamentos Intermedios/patología , Ratones , Neuronas/metabolismo , Médula Espinal/metabolismo
5.
JCI Insight ; 5(21)2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33021970

RESUMEN

Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD), 2 incurable neurodegenerative disorders, share the same pathological hallmark named TDP43 (TAR DNA binding protein 43) proteinopathy. This event is characterized by a consistent cytoplasmic mislocalization and aggregation of the protein TDP43, which loses its physiological properties, leading neurons to death. Antibody-based approaches are now emerging interventions in the field of neurodegenerative disorders. Here, we tested the target specificity, in vivo distribution, and therapeutic efficacy of a monoclonal full-length antibody, named E6, in TDP43-related conditions. We observed that the antibody recognizes specifically the cytoplasmic fraction of TDP43. We demonstrated its ability in targeting large neurons in the spinal cord of mice and in reducing TDP43 mislocalization and NF-κB activation. We also recognized the proteasome as well as the lysosome machineries as possible mechanisms used by the antibody to reduce TDP43 proteinopathy. To our knowledge, this is the first report showing the therapeutic efficacy and feasibility of a full-length antibody against TDP43 in reducing TDP43 proteinopathy in spinal neurons of an ALS/FTLD mouse model.


Asunto(s)
Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Anticuerpos Monoclonales/farmacología , Proteínas de Unión al ADN/inmunología , Neuronas/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Proteinopatías TDP-43/tratamiento farmacológico , Anciano , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , FN-kappa B/metabolismo , Neuronas/inmunología , Neuronas/patología , Médula Espinal/inmunología , Médula Espinal/patología , Proteinopatías TDP-43/inmunología , Proteinopatías TDP-43/metabolismo , Proteinopatías TDP-43/patología
6.
Neuroscience ; 405: 14-23, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29526689

RESUMEN

Microglia are the principle immune cells of the brain. Once activated, microglial cells may exhibit a wide repertoire of the context-dependent profiles ranging from highly neurotoxic to more protective and pro-regenerative cellular phenotypes. While to date the mechanisms involved in the molecular regulation of the microglia polarization phenotypes remain elusive, growing evidence suggests that gender may markedly affect the inflammatory and/or glial responses following brain injuries. In the recent years, special attention has been given to the role of microglia in sexual dimorphism, both in healthy brain and diseased brain. Here, we review recent advances revealing microglia as an important determinant of gender differences under physiological conditions and in injured brain. We also discuss how microglia-driven innate immunity and signaling pathways might be involved in the sex-dependent responses following brain ischemic injury. Finally we describe how advanced methods such as live imaging techniques may help elucidate the role of microglia in the modulation of immune responses and gender difference after stroke.


Asunto(s)
Lesiones Encefálicas/fisiopatología , Encéfalo/fisiopatología , Microglía/fisiología , Animales , Encéfalo/patología , Lesiones Encefálicas/patología , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Femenino , Humanos , Inmunidad Innata , Masculino , Microglía/patología , Globulina de Unión a Hormona Sexual
7.
Neurobiol Aging ; 40: 50-60, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26973103

RESUMEN

Estrogens are known to exert neuroprotective and immuneomodulatory effects after stroke. However, at present, little is known about the role of estrogens and its receptors in postischemic inflammation after menopause. Here, we provide important in vivo evidence of a distinct shift in microglial phenotypes in the model of postmenopause brain. Using a model-system for live imaging of microglial activation in the context of chronic estrogen- and ERα-deficiency associated with aging, we observed a marked deregulation of the TLR2 signals and/or microglial activation in ovariectomized and/or ERα knockout mice. Further analysis revealed a 5.7-fold increase in IL-6, a 4.7-fold increase in phospho-Stat3 levels suggesting an overactivation of JAK/STAT3 pathway and significantly larger infarction in ERα knockouts chronically deprived of estrogen. Taken together, our results suggest that in the experimental model of menopause and/or aging, ERα mediates innate immune responses and/or microglial activation, and ischemia-induced production of IL-6. Based on our results, we propose that the loss of functional ERα may lead to deregulation of postischemic inflammatory responses and increased vulnerability to ischemic injury in aging female brains.


Asunto(s)
Envejecimiento , Isquemia Encefálica/patología , Encéfalo/patología , Receptor alfa de Estrógeno/deficiencia , Receptor alfa de Estrógeno/fisiología , Menopausia , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Estrógenos/deficiencia , Estrógenos/fisiología , Femenino , Inmunidad Innata , Interleucina-6/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/patología , Modelos Animales , Factor de Transcripción STAT3/metabolismo
8.
Neurobiol Dis ; 66: 66-73, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24613658

RESUMEN

Post-ischemic inflammation plays an important role in the evolution of brain injury, recovery and repair after stroke. Housing rodents in an enriched environment provides multisensory stimulation to the brain and enhances functional recovery after experimental stroke, also depressing the release of cytokines and chemokines in the peri-infarct. In order to identify targets for late stroke treatment, we studied the dynamics of inflammation and the contribution of resident Toll-like receptor 2 (TLR2) expressing microglia cells. We took advantage of the biophotonic/bioluminescent imaging technique using the reporter mouse-expressing luciferase and GFP reporter genes under transcriptional control of the murine TLR2 promoter (TLR2-luc/GFP mice) for non-invasive in vivo analysis of TLR2 activation/response in photothrombotic stroke after differential housing. Real-time imaging at 1day after stroke, revealed up-regulation of TLR2 in response to photothrombotic stroke that subsequently declined over time of recovery (14days). The inflammatory response was persistently down-regulated within days of enriched housing, enhancing recovery of lost sensori-motor function in TLR2-luc mice without affecting infarct size. The number of YM1-expressing microglia in the peri-infarct and areas remote from the infarct was also markedly attenuated. Using a live imaging approach, we demonstrate that multisensory stimulation rapidly, persistently and generally attenuates brain inflammation after experimental stroke, reducing the TLR2 response and leading to improved neurological outcome. TLR2-expressing microglia cells may provide targets for new stroke therapeutics.


Asunto(s)
Encéfalo/metabolismo , Ambiente , Vivienda para Animales , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/terapia , Receptor Toll-Like 2/metabolismo , Animales , Encéfalo/patología , Recuento de Células , Regulación hacia Abajo , Encefalitis/metabolismo , Encefalitis/patología , Técnica del Anticuerpo Fluorescente , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica , Lectinas/metabolismo , Mediciones Luminiscentes , Masculino , Ratones , Ratones Transgénicos , Microglía/metabolismo , Recuperación de la Función , Accidente Cerebrovascular/patología , Receptor Toll-Like 2/genética , beta-N-Acetilhexosaminidasas/metabolismo
9.
J Neuroinflammation ; 9: 191, 2012 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-22873409

RESUMEN

BACKGROUND: Using a live imaging approach, we have previously shown that microglia activation after stroke is characterized by marked and long-term induction of the Toll-like receptor (TLR) 2 biophotonic signals. However, the role of TLR2 (and potentially other TLRs) beyond the acute innate immune response and as early neuroprotection against ischemic injury is not well understood. METHODS: TLR2-/- mice were subjected to transient middle cerebral artery occlusion followed by different reperfusion times. Analyses assessing microglial activation profile/innate immune response were performed using in situ hybridization, immunohistochemistry analysis, flow cytometry and inflammatory cytokine array. The effects of the TLR2 deficiency on the evolution of ischemic brain injury were analyzed using a cresyl violet staining of brain sections with appropriate lesion size estimation. RESULTS: Here we report that TLR2 deficiency markedly affects post-stroke immune response resulting in delayed exacerbation of the ischemic injury. The temporal analysis of the microglia/macrophage activation profiles in TLR2-/- mice and age-matched controls revealed reduced microglia/macrophage activation after stroke, reduced capacity of resident microglia to proliferate as well as decreased levels of monocyte chemotactic protein-1 (MCP-1) and consequently lower levels of CD45(high)/CD11b(+) expressing cells as shown by flow cytometry analysis. Importantly, although acute ischemic lesions (24 to 72 h) were smaller in TLR2-/- mice, the observed alterations in innate immune response were more pronounced at later time points (at day 7) after initial stroke, which finally resulted in delayed exacerbation of ischemic lesion leading to larger chronic infarctions as compared with wild-type mice. Moreover, our results revealed that TLR2 deficiency is associated with significant decrease in the levels of neurotrophic/anti-apoptotic factor Insulin-like growth factor-1 (IGF-1), expressed by microglia in the areas both in and around ischemic lesion. CONCLUSION: Our results clearly suggest that optimal and timely microglial activation/innate immune response is needed to limit neuronal damage after stroke.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Receptor Toll-Like 2/deficiencia , Animales , Isquemia Encefálica/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Microglía/patología , Factores de Tiempo , Receptor Toll-Like 2/biosíntesis , Receptor Toll-Like 2/genética
10.
Methods Enzymol ; 506: 117-33, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22341222

RESUMEN

Brain response to ischemic injury is characterized by initiation of a complex pathophysiological cascade comprising the events that may evolve over hours or several days and weeks after initial attack. At present, spatial and temporal dynamics of these events is not completely understood. To enable better understanding of the brain response to ischemic injury we developed and validated several novel transgenic mouse models of bioluminescence and fluorescence, allowing the noninvasive and time-lapse imaging of neuroinflammation, neuronal damage/stress and repair. These mice represent a powerful analytical tool for understanding in vivo pathology as well as the evaluating pharmacokinetics and longitudinal responses to drug therapies. Here, we describe the basic procedures of generating biophotonic mouse models for live imaging of microglial activation and neuronal stress and recovery, followed by a detailed description of in vivo bioluminescence imaging protocols used after experimental stroke.


Asunto(s)
Isquemia Encefálica/patología , Encéfalo/patología , Imagenología Tridimensional/métodos , Mediciones Luminiscentes/métodos , Neuronas/patología , Animales , Encéfalo/inmunología , Encéfalo/metabolismo , Isquemia Encefálica/genética , Isquemia Encefálica/inmunología , Modelos Animales de Enfermedad , Luciferina de Luciérnaga/análisis , Humanos , Sustancias Luminiscentes/análisis , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/inmunología , Neuronas/metabolismo
11.
Stroke ; 42(10): 2903-9, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21852616

RESUMEN

BACKGROUND AND PURPOSE: Consumption of fish has been shown to reduce risk of coronary heart disease and, possibly, of ischemic stroke. Because docosahexaenoic acid (DHA) is the most likely neuroactive component within fish oil, we hypothesized that exposing mice to a DHA-enriched diet may reduce inflammation and protect neurons against ischemic injury. METHODS: To visualize the effects of DHA on neuroinflammation after stroke, TLR2-fluc-GFP transgenic mice were exposed to either a control diet, a diet depleted in n-3 polyunsaturated fatty acid, or a diet enriched in DHA during 3 months. Real-time biophotonic/bioluminescence imaging of the TLR2 response was performed before and after middle cerebral artery occlusion, whereas cytokines concentrations and stroke area analyses were performed at 3 and 7 days after middle cerebral artery occlusion, respectively. RESULTS: We show that a 3-month DHA treatment prevented microglial activation after ischemic injury, reduced the ischemic lesion size, and increased levels of the antiapoptotic molecule Bcl-2 in the brain. Additional analysis revealed a significant decrease in the levels of COX2 and IL-1ß, but not in other proinflammatory cytokines. Importantly, long-term DHA supplementation significantly changed the n-3:n-6 polyunsaturated fatty acid ratio in the brain. CONCLUSIONS: Collectively, these data indicate that diet-induced accumulation of DHA in the brain protects against postischemic inflammation and injury. Because DHA is widely available at low cost and has an excellent safety profile, our data suggest that increased DHA intake may provide protection against acute immune response/brain damage in ischemic stroke.


Asunto(s)
Isquemia Encefálica/inmunología , Encéfalo/inmunología , Ácidos Docosahexaenoicos/administración & dosificación , Inmunidad Activa/inmunología , Neuronas/inmunología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Ácidos Docosahexaenoicos/metabolismo , Inflamación/inmunología , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Receptor Toll-Like 2/metabolismo
12.
Stroke ; 39(3): 935-42, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18258827

RESUMEN

BACKGROUND AND PURPOSE: We sought to develop a model system for live analysis of brain inflammatory response in ischemic injury. METHODS: Using a reporter mouse-expressing luciferase gene under transcriptional control of the murine glial fibrillary acidic protein (GFAP) promoter (GFAP-luc mice) and biophotonic/bioluminescent imaging as tools, we developed a model system for in vivo analysis of astrocyte activation/response in cerebral ischemia. RESULTS: Analysis of photon emissions from the brains of living animals revealed marked sex differences in astrocyte response to ischemic injury. The increase in GFAP signals was significantly higher in female mice in the metestrus/diestrus period compared with male transgenic mice (1.71 x 10(7)+/-0.19 x 10(7) vs 0.92 x 10(7)+/-0.15 x 10(7), P<0.001). Similar results were obtained by quantitative immunohistochemistry (males vs females: 13.4+/-0.5 vs 16.96+/-0.64, P<0.0001). However, astrocyte activation/GFAP signals showed cyclic, estrus-dependent variations in response to ischemic injury. Physiologically higher levels of estrogen and application of pharmacologic doses of estrogen during replacement therapy attenuated GFAP upregulation after stroke. Interestingly, contrary to a positive correlation between the intensities of GFAP signals and infarct size in male mice, no such correlation was observed in any of the experimental groups of female GFAP-luc mice. CONCLUSIONS: Our results suggest that GFAP upregulation in ischemic injury may have different functional significance in female and male experimental animals and may not directly reflect the extent of ischemia-induced neuronal damage in female GFAP-luc mice. Using a novel live imaging approach, we demonstrated that the early-phase brain inflammatory response to ischemia may be associated with sex-specific biomarkers of brain damage.


Asunto(s)
Astrocitos/patología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Encefalitis/diagnóstico , Estrógenos/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Factores Sexuales , Animales , Isquemia Encefálica/complicaciones , Infarto Cerebral/etiología , Infarto Cerebral/patología , Diestro , Encefalitis/etiología , Encefalitis/metabolismo , Estrógenos/farmacología , Femenino , Proteína Ácida Fibrilar de la Glía/genética , Inmunohistoquímica , Luminiscencia , Masculino , Metestro , Ratones , Ratones Transgénicos , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...