Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Cell ; 185(22): 4135-4152.e22, 2022 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-36257314

RESUMEN

Recent studies have begun to reveal critical roles for the brain's professional phagocytes, microglia, and their receptors in the control of neurotoxic amyloid beta (Aß) and myelin debris accumulation in neurodegenerative disease. However, the critical intracellular molecules that orchestrate neuroprotective functions of microglia remain poorly understood. In our studies, we find that targeted deletion of SYK in microglia leads to exacerbated Aß deposition, aggravated neuropathology, and cognitive defects in the 5xFAD mouse model of Alzheimer's disease (AD). Disruption of SYK signaling in this AD model was further shown to impede the development of disease-associated microglia (DAM), alter AKT/GSK3ß-signaling, and restrict Aß phagocytosis by microglia. Conversely, receptor-mediated activation of SYK limits Aß load. We also found that SYK critically regulates microglial phagocytosis and DAM acquisition in demyelinating disease. Collectively, these results broaden our understanding of the key innate immune signaling molecules that instruct beneficial microglial functions in response to neurotoxic material.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Animales , Ratones , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides , Modelos Animales de Enfermedad , Ratones Transgénicos , Microglía/patología , Fagocitosis
2.
Nature ; 580(7805): 647-652, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32350463

RESUMEN

Neurodevelopment is characterized by rapid rates of neural cell proliferation and differentiation followed by massive cell death in which more than half of all recently generated brain cells are pruned back. Large amounts of DNA damage, cellular debris, and by-products of cellular stress are generated during these neurodevelopmental events, all of which can potentially activate immune signalling. How the immune response to this collateral damage influences brain maturation and function remains unknown. Here we show that the AIM2 inflammasome contributes to normal brain development and that disruption of this immune sensor of genotoxic stress leads to behavioural abnormalities. During infection, activation of the AIM2 inflammasome in response to double-stranded DNA damage triggers the production of cytokines as well as a gasdermin-D-mediated form of cell death known as pyroptosis1-4. We observe pronounced AIM2 inflammasome activation in neurodevelopment and find that defects in this sensor of DNA damage result in anxiety-related behaviours in mice. Furthermore, we show that the AIM2 inflammasome contributes to central nervous system (CNS) homeostasis specifically through its regulation of gasdermin-D, and not via its involvement in the production of the cytokines IL-1 and/or IL-18. Consistent with a role for this sensor of genomic stress in the purging of genetically compromised CNS cells, we find that defective AIM2 inflammasome signalling results in decreased neural cell death both in response to DNA damage-inducing agents and during neurodevelopment. Moreover, mutations in AIM2 lead to excessive accumulation of DNA damage in neurons as well as an increase in the number of neurons that incorporate into the adult brain. Our findings identify the inflammasome as a crucial player in establishing a properly formed CNS through its role in the removal of genetically compromised cells.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Inflamasomas/metabolismo , Animales , Animales Recién Nacidos , Ansiedad/patología , Ansiedad/fisiopatología , Ansiedad/psicología , Conducta Animal/fisiología , Encéfalo/citología , Encéfalo/metabolismo , Encéfalo/patología , Proteínas Adaptadoras de Señalización CARD/metabolismo , Caspasa 1/deficiencia , Caspasa 1/metabolismo , Muerte Celular , Proteínas de Unión al ADN/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Mutación , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neuronas/citología , Neuronas/metabolismo , Neuronas/patología , Proteínas de Unión a Fosfato/metabolismo
3.
Mol Psychiatry ; 28(3): 1101-1111, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36481930

RESUMEN

We developed an IGFBP2-mimetic peptide fragment, JB2, and showed that it promotes basal synaptic structural and functional plasticity in cultured neurons and mice. We demonstrate that JB2 directly binds to dendrites and synapses, and its biological activity involves NMDA receptor activation, gene transcription and translation, and IGF2 receptors. It is not IGF1 receptor-dependent. In neurons, JB2 induced extensive remodeling of the membrane phosphoproteome. Synapse and cytoskeletal regulation, autism spectrum disorder (ASD) risk factors, and a Shank3-associated protein network were significantly enriched among phosphorylated and dephosphorylated proteins. Haploinsufficiency of the SHANK3 gene on chromosome 22q13.3 often causes Phelan-McDermid Syndrome (PMS), a genetically defined form of autism with profound deficits in motor behavior, sensory processing, language, and cognitive function. We identified multiple disease-relevant phenotypes in a Shank3 heterozygous mouse and showed that JB2 rescued deficits in synaptic function and plasticity, learning and memory, ultrasonic vocalizations, and motor function; it also normalized neuronal excitability and seizure susceptibility. Notably, JB2 rescued deficits in the auditory evoked response latency, alpha peak frequency, and steady-state electroencephalography response, measures with direct translational value to human subjects. These data demonstrate that JB2 is a potent modulator of neuroplasticity with therapeutic potential for the treatment of PMS and ASD.


Asunto(s)
Trastorno del Espectro Autista , Trastornos de los Cromosomas , Humanos , Ratones , Animales , Trastorno del Espectro Autista/genética , Proteínas del Tejido Nervioso/genética , Deleción Cromosómica , Trastornos de los Cromosomas/genética , Péptidos/genética , Modelos Animales de Enfermedad , Plasticidad Neuronal , Cromosomas Humanos Par 22/metabolismo , Proteínas de Microfilamentos/genética
4.
Brain Behav Immun ; 108: 80-97, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36343752

RESUMEN

Perturbations to the in utero environment can dramatically change the trajectory of offspring neurodevelopment. Insults commonly encountered in modern human life such as infection, toxins, high-fat diet, prescription medications, and others are increasingly linked to behavioral alterations in prenatally-exposed offspring. While appreciation is expanding for the potential consequence that these triggers can have on embryo development, there is a paucity of information concerning how the crucial maternal-fetal interface (MFI) responds to these various insults and how it may relate to changes in offspring neurodevelopment. Here, we found that the MFI responds both to an inflammatory state and altered serotonergic tone in pregnant mice. Maternal immune activation (MIA) triggered an acute inflammatory response in the MFI dominated by interferon signaling that came at the expense of ordinary development-related transcriptional programs. The major MFI compartments, the decidua and the placenta, each responded in distinct manners to MIA. MFIs exposed to MIA were also found to have disrupted sex-specific gene expression and heightened serotonin levels. We found that offspring exposed to MIA had sex-biased behavioral changes and that microglia were not transcriptionally impacted. Moreover, the combination of maternal inflammation in the presence of pharmacologic inhibition of serotonin reuptake further transformed MFI physiology and offspring neurobiology, impacting immune and serotonin signaling pathways alike. In all, these findings highlight the complexities of evaluating diverse environmental impacts on placental physiology and neurodevelopment.


Asunto(s)
Placenta , Efectos Tardíos de la Exposición Prenatal , Masculino , Embarazo , Ratones , Animales , Femenino , Humanos , Placenta/metabolismo , Serotonina/metabolismo , Neurobiología , Inflamación/metabolismo
5.
RNA Biol ; 17(8): 1183-1195, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31983265

RESUMEN

tRNA-derived small fragments (tRFs) and tRNA halves have emerging functions in different biological pathways, such as regulating gene expression, protein translation, retrotransposon activity, transgenerational epigenetic changes and response to environmental stress. However, small RNAs like tRFs and microRNAs in the maternal-fetal interface during gestation have not been studied extensively. Here we investigated the small RNA composition of mouse placenta/decidua, which represents the interface where the mother communicates with the foetus, to determine whether there are specific differences in tRFs and microRNAs during fetal development and in response to maternal immune activation (MIA). Global tRF expression pattern, just like microRNAs, can distinguish tissue types among placenta/decidua, fetal brain and fetal liver. In particular, 5' tRNA halves from tRNAGly, tRNAGlu, tRNAVal and tRNALys are abundantly expressed in the normal mouse placenta/decidua. Moreover, tRF and microRNA levels in the maternal-fetal interface change dynamically over the course of embryonic development. To see if stress alters non-coding RNA expression at the maternal-fetal interface, we treated pregnant mice with a viral infection mimetic, which has been shown to promote autism-related phenotypes in the offspring. Acute changes in the levels of specific tRFs and microRNAs were observed 3-6 h after MIA and are suppressed thereafter. A group of 5' tRNA halves is down-regulated by MIA, whereas a group of 18-nucleotide tRF-3a is up-regulated. In conclusion, tRFs show tissue-specificity, developmental changes and acute response to environmental stress, opening the possibility of them having a role in the fetal response to MIA.


Asunto(s)
Trastorno Autístico/etiología , MicroARNs/genética , Placenta/metabolismo , ARN Pequeño no Traducido/genética , ARN de Transferencia/genética , Animales , Trastorno Autístico/metabolismo , Trastorno Autístico/psicología , Decidua/metabolismo , Femenino , Regulación de la Expresión Génica , Ratones , MicroARNs/metabolismo , Embarazo , ARN Pequeño no Traducido/metabolismo , ARN de Transferencia/metabolismo
6.
J Immunol ; 201(3): 845-850, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29967099

RESUMEN

Recent studies suggest that autism is often associated with dysregulated immune responses and altered microbiota composition. This has led to growing speculation about potential roles for hyperactive immune responses and the microbiome in autism. Yet how microbiome-immune cross-talk contributes to neurodevelopmental disorders currently remains poorly understood. In this study, we report critical roles for prenatal microbiota composition in the development of behavioral abnormalities in a murine maternal immune activation (MIA) model of autism that is driven by the viral mimetic polyinosinic-polycytidylic acid. We show that preconception microbiota transplantation can transfer susceptibility to MIA-associated neurodevelopmental disease and that this is associated with modulation of the maternal immune response. Furthermore, we find that ablation of IL-17a signaling provides protection against the development of neurodevelopmental abnormalities in MIA offspring. Our findings suggest that microbiota landscape can influence MIA-induced neurodevelopmental disease pathogenesis and that this occurs as a result of microflora-associated calibration of gestational IL-17a responses.


Asunto(s)
Trastorno Autístico/inmunología , Trastorno Autístico/microbiología , Sistema Inmunológico/inmunología , Microbiota/inmunología , Efectos Tardíos de la Exposición Prenatal/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Interleucina-17/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Poli I-C/inmunología , Embarazo , Efectos Tardíos de la Exposición Prenatal/microbiología
7.
Hum Mol Genet ; 25(1): 130-45, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26566673

RESUMEN

Genetic background significantly affects phenotype in multiple mouse models of human diseases, including muscular dystrophy. This phenotypic variability is partly attributed to genetic modifiers that regulate the disease process. Studies have demonstrated that introduction of the γ-sarcoglycan-null allele onto the DBA/2J background confers a more severe muscular dystrophy phenotype than the original strain, demonstrating the presence of genetic modifier loci in the DBA/2J background. To characterize the phenotype of dystrophin deficiency on the DBA/2J background, we created and phenotyped DBA/2J-congenic Dmdmdx mice (D2-mdx) and compared them with the original, C57BL/10ScSn-Dmdmdx (B10-mdx) model. These strains were compared with their respective control strains at multiple time points between 6 and 52 weeks of age. Skeletal and cardiac muscle function, inflammation, regeneration, histology and biochemistry were characterized. We found that D2-mdx mice showed significantly reduced skeletal muscle function as early as 7 weeks and reduced cardiac function by 28 weeks, suggesting that the disease phenotype is more severe than in B10-mdx mice. In addition, D2-mdx mice showed fewer central myonuclei and increased calcifications in the skeletal muscle, heart and diaphragm at 7 weeks, suggesting that their pathology is different from the B10-mdx mice. The new D2-mdx model with an earlier onset and more pronounced dystrophy phenotype may be useful for evaluating therapies that target cardiac and skeletal muscle function in dystrophin-deficient mice. Our data align the D2-mdx with Duchenne muscular dystrophy patients with the LTBP4 genetic modifier, making it one of the few instances of cross-species genetic modifiers of monogenic traits.


Asunto(s)
Modelos Animales de Enfermedad , Antecedentes Genéticos , Distrofia Muscular Animal/genética , Animales , Peso Corporal , Distrofina/genética , Ecocardiografía , Femenino , Fuerza de la Mano , Pruebas de Función Cardíaca , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Endogámicos mdx , Contracción Muscular , Músculos/patología , Distrofia Muscular Animal/patología , Miofibrillas/patología , Miositis/genética , Miositis/patología , Tamaño de los Órganos , Fenotipo
8.
Artículo en Inglés | MEDLINE | ID: mdl-37520631

RESUMEN

This special edition is based on the revelation that "the lessons learned and unlearned during COVID-19 grant us an unparalleled opportunity to reflect." Here, we reflect on lessons learned related to teacher adaptiveness. We examined how the COVID-19 pandemic demonstrated the adaptiveness necessary for teachers to knowledge generation approaches aligned with the Next Generation Science Standards. First, we outline a three-year professional development program focused on knowledge generation approaches. We present findings from teachers' experiences teaching science from 2019 to 2021, collected through consecutive form explanatory mixed-methods analysis involving written responses to vignettes (n = 474) and classroom observations (n = 58). Then, using an individual teacher case study, we explore how the shift to virtual teaching was supported by adaptiveness. Results suggest a significant relationship between teacher adaptiveness and the use of knowledge generation approaches. We conclude with implications for elementary science teacher professional development and present questions for further research on adaptiveness. Supplementary Information: The online version contains supplementary material available at 10.1186/s43031-022-00052-3.

9.
Nat Commun ; 11(1): 4524, 2020 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-32913280

RESUMEN

Traumatic brain injury (TBI) is a leading global cause of death and disability. Here we demonstrate in an experimental mouse model of TBI that mild forms of brain trauma cause severe deficits in meningeal lymphatic drainage that begin within hours and last out to at least one month post-injury. To investigate a mechanism underlying impaired lymphatic function in TBI, we examined how increased intracranial pressure (ICP) influences the meningeal lymphatics. We demonstrate that increased ICP can contribute to meningeal lymphatic dysfunction. Moreover, we show that pre-existing lymphatic dysfunction before TBI leads to increased neuroinflammation and negative cognitive outcomes. Finally, we report that rejuvenation of meningeal lymphatic drainage function in aged mice can ameliorate TBI-induced gliosis. These findings provide insights into both the causes and consequences of meningeal lymphatic dysfunction in TBI and suggest that therapeutics targeting the meningeal lymphatic system may offer strategies to treat TBI.


Asunto(s)
Lesiones Encefálicas/fisiopatología , Gliosis/fisiopatología , Sistema Glinfático/fisiología , Meninges/fisiopatología , Animales , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/patología , Lesiones Encefálicas/terapia , Dependovirus/genética , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Gliosis/etiología , Gliosis/patología , Gliosis/prevención & control , Sistema Glinfático/patología , Humanos , Masculino , Meninges/patología , Ratones , Factor C de Crecimiento Endotelial Vascular/genética , Factor C de Crecimiento Endotelial Vascular/uso terapéutico
10.
Methods Mol Biol ; 1960: 227-236, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30798536

RESUMEN

Autism spectrum disorder (ASD) has emerged as one of the most prevalent and poorly understood disorders of our time. The etiology of autism currently remains poorly understood; however, emerging clinical and experimental evidence suggests central roles for maternal immune activation (MIA) during pregnancy in ASD. In particular, children whose mothers suffered from an infectious disease or other inflammatory conditions during pregnancy are at a substantially higher risk of developing ASD. It has been shown that MIA-induced ASD can be modeled by treating pregnant mice with the viral mimetic polyinosinic-polycytidylic acid (PolyI:C) during key neurodevelopmental time points. In this paradigm, PolyI:C treatment induces systemic inflammatory responses that model MIA during viral infections. Offspring from PolyI:C-treated mothers develop many of the defining features of ASD including defects in social interactions, communicative impairments, and repetitive/stereotyped behaviors, as well as neuropathologies that are commonly observed in human ASD. While the early use of this emerging ASD model system has provided important initial insights into the involvement of gestational immune dysfunction in neurodevelopmental disorder pathogenesis, we have only just begun to scratch the surface in our understanding of how MIA affects brain maturation and contributes to neurodevelopmental disease. Here we describe best practices for how the PolyI:C model of MIA can be used to study autism-related disorders in mice.


Asunto(s)
Trastorno del Espectro Autista/inmunología , Microbiota/fisiología , Animales , Trastorno del Espectro Autista/inducido químicamente , Conducta Animal , Modelos Animales de Enfermedad , Femenino , Ratones , Poli I-C/farmacología , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA