Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Nature ; 609(7929): 1021-1028, 2022 09.
Article in English | MEDLINE | ID: mdl-36131014

ABSTRACT

Medulloblastoma (MB) comprises a group of heterogeneous paediatric embryonal neoplasms of the hindbrain with strong links to early development of the hindbrain1-4. Mutations that activate Sonic hedgehog signalling lead to Sonic hedgehog MB in the upper rhombic lip (RL) granule cell lineage5-8. By contrast, mutations that activate WNT signalling lead to WNT MB in the lower RL9,10. However, little is known about the more commonly occurring group 4 (G4) MB, which is thought to arise in the unipolar brush cell lineage3,4. Here we demonstrate that somatic mutations that cause G4 MB converge on the core binding factor alpha (CBFA) complex and mutually exclusive alterations that affect CBFA2T2, CBFA2T3, PRDM6, UTX and OTX2. CBFA2T2 is expressed early in the progenitor cells of the cerebellar RL subventricular zone in Homo sapiens, and G4 MB transcriptionally resembles these progenitors but are stalled in developmental time. Knockdown of OTX2 in model systems relieves this differentiation blockade, which allows MB cells to spontaneously proceed along normal developmental differentiation trajectories. The specific nature of the split human RL, which is destined to generate most of the neurons in the human brain, and its high level of susceptible EOMES+KI67+ unipolar brush cell progenitor cells probably predisposes our species to the development of G4 MB.


Subject(s)
Cell Differentiation , Cerebellar Neoplasms , Medulloblastoma , Metencephalon , Cell Differentiation/genetics , Cell Lineage , Cerebellar Neoplasms/classification , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Cerebellum/embryology , Cerebellum/pathology , Core Binding Factor alpha Subunits/genetics , Hedgehog Proteins/metabolism , Histone Demethylases , Humans , Ki-67 Antigen/metabolism , Medulloblastoma/classification , Medulloblastoma/genetics , Medulloblastoma/pathology , Metencephalon/embryology , Metencephalon/pathology , Muscle Proteins , Mutation , Otx Transcription Factors/deficiency , Otx Transcription Factors/genetics , Repressor Proteins , T-Box Domain Proteins/metabolism , Transcription Factors
2.
NMR Biomed ; : e5222, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164196

ABSTRACT

Microglia have been shown to proliferate and become activated following cranial radiotherapy (CRT), resulting in a chronic inflammatory response. We investigated the role of microglia in contributing to widespread volume losses observed in the brain following CRT in juvenile mice. To manipulate microglia, we used low-dose treatment with a highly selective CSF1R inhibitor called PLX5622 (PLX). We hypothesized that alteration of the post-CRT microglia population would lead to changes in brain development outcomes, as evaluated by structural MRI. Wild-type C57BL/6J mice were provided with daily intraperitoneal injections of PLX (25 mg/kg) or vehicle from postnatal day (P)14 to P19. Mice also received whole-brain irradiation (7 Gy) or sham irradiation (0 Gy) at 16 days of age. In one cohort of mice, immunohistochemical assessment in tissue sections was conducted to assess the impact of the selected PLX and CRT doses as well as their combination. In a separate cohort, mice were imaged using MRI at P14 (pretreatment), P19, P23, P42 and P63 in order to assess induced volume changes, which were measured based on structures from a predefined atlas. We observed that PLX and radiation treatments led to sex-specific changes in the microglial cell population. Across treatment groups, MRI-detected anatomical volumes at P19 and P63 were associated with microglia and proliferating microglia densities, respectively. Overall, our study demonstrates that low-dose PLX treatment produces a sex-dependent response in juvenile mice, that manipulation of microglia alters CRT-induced volume changes and that microglia density and MRI-derived volume changes are correlated in this model.

3.
Brain Behav Immun ; 119: 637-647, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38663773

ABSTRACT

Obesity is a major modifiable risk factor for Alzheimer's disease (AD), characterized by progressive atrophy of the cerebral cortex. The neurobiology of obesity contributions to AD is poorly understood. Here we show with in vivo MRI that diet-induced obesity decreases cortical volume in mice, and that higher body adiposity associates with lower cortical volume in humans. Single-nuclei transcriptomics of the mouse cortex reveals that dietary obesity promotes an array of neuron-adverse transcriptional dysregulations, which are mediated by an interplay of excitatory neurons and glial cells, and which involve microglial activation and lowered neuronal capacity for neuritogenesis and maintenance of membrane potential. The transcriptional dysregulations of microglia, more than of other cell types, are like those in AD, as assessed with single-nuclei cortical transcriptomics in a mouse model of AD and two sets of human donors with the disease. Serial two-photon tomography of microglia demonstrates microgliosis throughout the mouse cortex. The spatial pattern of adiposity-cortical volume associations in human cohorts interrogated together with in silico bulk and single-nucleus transcriptomic data from the human cortex implicated microglia (along with other glial cells and subtypes of excitatory neurons), and it correlated positively with the spatial profile of cortical atrophy in patients with mild cognitive impairment and AD. Thus, multi-cell neuron-adverse dysregulations likely contribute to the loss of cortical tissue in obesity. The dysregulations of microglia may be pivotal to the obesity-related risk of AD.


Subject(s)
Alzheimer Disease , Cerebral Cortex , Obesity , Animals , Obesity/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Humans , Mice , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Male , Microglia/metabolism , Neurons/metabolism , Female , Disease Models, Animal , Mice, Inbred C57BL , Magnetic Resonance Imaging , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/etiology , Atrophy , Diet, High-Fat/adverse effects , Aged , Adiposity , Transcriptome
5.
Anim Cogn ; 25(6): 1599-1608, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35731425

ABSTRACT

Serial reaction time tasks, in which subjects have to match a target to a cue, are used to explore whether non-human animals have multiple memory systems. Predictable sub-sequences embedded in the sequence of cues are responded to faster, demonstrating incidental learning, often considered implicit. Here, we used the serial implicit learning task (SILT) to determine whether rats' memory shows similar effects. In SILT, subjects must nose-poke into a sequence of two lit apertures, S1 and S2. Some S1 are always followed by the same S2, creating predictable sequences (PS). Across groups, we varied the proportion of PS trials, from 10 to 80%, and show that rats with more PS experience do better on them than on unpredictable sequences, and better than rats with less experience. We then introduced test trials in which no S2 was cued. Rats with more PS experience did better on test trials. Finally, we reversed some sequences (from predictable to unpredictable and vice versa) and changed others. We find that rats with more PS experience perseverate on old (now incorrect) responses more than those with less PS experience. Overall, we find a discontinuity in performance as the proportion of PS increases, suggesting a switch in behavioral strategies or memory systems, which we confirm using a Process Dissociation Procedure analysis. Our data suggest that rats have at least two distinct memory systems, one of which appears to be analogous to human implicit memory and is differentially activated by varying the proportion of PS in our task.


Subject(s)
Learning , Serial Learning , Rats , Humans , Animals , Reaction Time/physiology , Serial Learning/physiology , Learning/physiology , Cues
6.
Radiat Res ; 196(4): 394-403, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34270782

ABSTRACT

Sequelae after pediatric cranial radiotherapy (CRT) result in long-term changes in brain structure. While past evidence indicates regional differences in brain volume change, it remains unclear how these manifest in the time course of change after CRT. In this study, we spatiotemporally characterized volume losses induced by cranial irradiation in a mouse model, with a dense sampling of measurements over the first week postirradiation. Wild-type mice received whole-brain irradiation (7 Gy) or sham irradiation (0 Gy) at 16 days of age. In vivo magnetic resonance imaging was performed at one time point before, and 2-4 time points postirradiation in each mouse, with a particular focus on sampling during the first week after cranial irradiation. Volume changes across the brain were measured, and the degree and timing of volume loss were quantified across structures from a predefined atlas. Volume measurements across the brain after cranial irradiation revealed a ∼2-day delay in which volume is not significantly altered, after which time volume change proceeds over the course of four days. Volume losses were 3% larger and emerged 40% slower in white matter than in gray matter. Large volume loss was also observed in the ventricles. Differences in the timing and magnitude of volume change between gray and white matter after cranial irradiation were observed. These results suggest differences in the mechanism and/or kinetics underlying the associated radio-response, which may have implications in development.


Subject(s)
Cranial Irradiation , Animals , Brain , Mice , Mice, Inbred C57BL
7.
Nat Med ; 26(8): 1285-1294, 2020 08.
Article in English | MEDLINE | ID: mdl-32719487

ABSTRACT

We asked whether pharmacological stimulation of endogenous neural precursor cells (NPCs) may promote cognitive recovery and brain repair, focusing on the drug metformin, in parallel rodent and human studies of radiation injury. In the rodent cranial radiation model, we found that metformin enhanced the recovery of NPCs in the dentate gyrus, with sex-dependent effects on neurogenesis and cognition. A pilot double-blind, placebo-controlled crossover trial was conducted (ClinicalTrials.gov, NCT02040376) in survivors of pediatric brain tumors who had been treated with cranial radiation. Safety, feasibility, cognitive tests and MRI measures of white matter and the hippocampus were evaluated as endpoints. Twenty-four participants consented and were randomly assigned to complete 12-week cycles of metformin (A) and placebo (B) in either an AB or BA sequence with a 10-week washout period at crossover. Blood draws were conducted to monitor safety. Feasibility was assessed as recruitment rate, medication adherence and procedural adherence. Linear mixed modeling was used to examine cognitive and MRI outcomes as a function of cycle, sequence and treatment. We found no clinically relevant safety concerns and no serious adverse events associated with metformin. Sequence effects were observed for all cognitive outcomes in our linear mixed models. For the subset of participants with complete data in cycle 1, metformin was associated with better performance than placebo on tests of declarative and working memory. We present evidence that a clinical trial examining the effects of metformin on cognition and brain structure is feasible in long-term survivors of pediatric brain tumors and that metformin is safe to use and tolerable in this population. This pilot trial was not intended to test the efficacy of metformin for cognitive recovery and brain growth, but the preliminary results are encouraging and warrant further investigation in a large multicenter phase 3 trial.


Subject(s)
Brain Neoplasms/complications , Cognitive Dysfunction/drug therapy , Metformin/administration & dosage , Pediatrics/trends , Adolescent , Adult , Brain/diagnostic imaging , Brain/drug effects , Brain/pathology , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cancer Survivors , Child , Child, Preschool , Cognition/drug effects , Cognitive Dysfunction/etiology , Cognitive Dysfunction/pathology , Double-Blind Method , Female , Humans , Magnetic Resonance Imaging , Male , Metformin/adverse effects , Neurogenesis/drug effects , Pilot Projects , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL