Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Sci Adv ; 8(37): eabo7639, 2022 09 16.
Article in English | MEDLINE | ID: mdl-36112685

ABSTRACT

To maintain homeostasis, the body, including the brain, reprograms its metabolism in response to altered nutrition or disease. However, the consequences of these challenges for the energy metabolism of the different brain cell types remain unknown. Here, we generated a proteome atlas of the major central nervous system (CNS) cell types from young and adult mice, after feeding the therapeutically relevant low-carbohydrate, high-fat ketogenic diet (KD) and during neuroinflammation. Under steady-state conditions, CNS cell types prefer distinct modes of energy metabolism. Unexpectedly, the comparison with KD revealed distinct cell type-specific strategies to manage the altered availability of energy metabolites. Astrocytes and neurons but not oligodendrocytes demonstrated metabolic plasticity. Moreover, inflammatory demyelinating disease changed the neuronal metabolic signature in a similar direction as KD. Together, these findings highlight the importance of the metabolic cross-talk between CNS cells and between the periphery and the brain to manage altered nutrition and neurological disease.


Subject(s)
Brain , Diet, Ketogenic , Animals , Brain/metabolism , Carbohydrates , Ketone Bodies/metabolism , Mice , Proteome/metabolism
2.
Front Neurosci ; 16: 917071, 2022.
Article in English | MEDLINE | ID: mdl-36061596

ABSTRACT

Direct neuronal reprogramming, the process whereby a terminally differentiated cell is converted into an induced neuron without traversing a pluripotent state, has tremendous therapeutic potential for a host of neurodegenerative diseases. While there is strong evidence for astrocyte-to-neuron conversion in vitro, in vivo studies in the adult brain are less supportive or controversial. Here, we set out to enhance the efficacy of neuronal conversion of adult astrocytes in vivo by optimizing the neurogenic capacity of a driver transcription factor encoded by the proneural gene Ascl1. Specifically, we mutated six serine phospho-acceptor sites in Ascl1 to alanines (Ascl1 SA 6) to prevent phosphorylation by proline-directed serine/threonine kinases. Native Ascl1 or Ascl1 SA 6 were expressed in adult, murine cortical astrocytes under the control of a glial fibrillary acidic protein (GFAP) promoter using adeno-associated viruses (AAVs). When targeted to the cerebral cortex in vivo, mCherry+ cells transduced with AAV8-GFAP-Ascl1 SA 6-mCherry or AAV8-GFAP-Ascl1-mCherry expressed neuronal markers within 14 days post-transduction, with Ascl1 SA 6 promoting the formation of more mature dendritic arbors compared to Ascl1. However, mCherry expression disappeared by 2-months post-transduction of the AAV8-GFAP-mCherry control-vector. To circumvent reporter issues, AAV-GFAP-iCre (control) and AAV-GFAP-Ascl1 (or Ascl1 SA 6)-iCre constructs were generated and injected into the cerebral cortex of Rosa reporter mice. In all comparisons of AAV capsids (AAV5 and AAV8), GFAP promoters (long and short), and reporter mice (Rosa-zsGreen and Rosa-tdtomato), Ascl1 SA 6 transduced cells more frequently expressed early- (Dcx) and late- (NeuN) neuronal markers. Furthermore, Ascl1 SA 6 repressed the expression of astrocytic markers Sox9 and GFAP more efficiently than Ascl1. Finally, we co-transduced an AAV expressing ChR2-(H134R)-YFP, an optogenetic actuator. After channelrhodopsin photostimulation, we found that Ascl1 SA 6 co-transduced astrocytes exhibited a significantly faster decay of evoked potentials to baseline, a neuronal feature, when compared to iCre control cells. Taken together, our findings support an enhanced neuronal conversion efficiency of Ascl1 SA 6 vs. Ascl1, and position Ascl1 SA 6 as a critical transcription factor for future studies aimed at converting adult brain astrocytes to mature neurons to treat disease.

3.
PLoS Biol ; 18(11): e3000943, 2020 11.
Article in English | MEDLINE | ID: mdl-33196637

ABSTRACT

In several neurodegenerative disorders, axonal pathology may originate from impaired oligodendrocyte-to-axon support of energy substrates. We previously established transgenic mice that allow measuring axonal ATP levels in electrically active optic nerves. Here, we utilize this technique to explore axonal ATP dynamics in the Plpnull/y mouse model of spastic paraplegia. Optic nerves from Plpnull/y mice exhibited lower and more variable basal axonal ATP levels and reduced compound action potential (CAP) amplitudes, providing a missing link between axonal pathology and a role of oligodendrocytes in brain energy metabolism. Surprisingly, when Plpnull/y optic nerves are challenged with transient glucose deprivation, both ATP levels and CAP decline slower, but recover faster upon reperfusion of glucose. Structurally, myelin sheaths display an increased frequency of cytosolic channels comprising glucose and monocarboxylate transporters, possibly facilitating accessibility of energy substrates to the axon. These data imply that complex metabolic alterations of the axon-myelin unit contribute to the phenotype of Plpnull/y mice.


Subject(s)
Adenosine Triphosphate/metabolism , Myelin Sheath/metabolism , Paraplegia/metabolism , Action Potentials , Animals , Axons/metabolism , Disease Models, Animal , Energy Metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Electron, Transmission , Microscopy, Immunoelectron , Myelin Proteolipid Protein/deficiency , Myelin Proteolipid Protein/genetics , Myelin Sheath/pathology , Optic Nerve/metabolism , Optic Nerve/pathology , Paraplegia/genetics , Paraplegia/pathology , Phenotype
4.
Nat Commun ; 11(1): 5497, 2020 10 30.
Article in English | MEDLINE | ID: mdl-33127910

ABSTRACT

Myelinating oligodendrocytes enable fast propagation of action potentials along the ensheathed axons. In addition, oligodendrocytes play diverse non-canonical roles including axonal metabolic support and activity-dependent myelination. An open question remains whether myelination also contributes to information processing in addition to speeding up conduction velocity. Here, we analyze the role of myelin in auditory information processing using paradigms that are also good predictors of speech understanding in humans. We compare mice with different degrees of dysmyelination using acute multiunit recordings in the auditory cortex, in combination with behavioral readouts. We find complex alterations of neuronal responses that reflect fatigue and temporal acuity deficits. We observe partially discriminable but similar deficits in well myelinated mice in which glial cells cannot fully support axons metabolically. We suggest a model in which myelination contributes to sustained stimulus perception in temporally complex paradigms, with a role of metabolically active oligodendrocytes in cortical information processing.


Subject(s)
Axons/metabolism , Myelin Sheath/metabolism , Oligodendroglia/physiology , Action Potentials/physiology , Animals , Auditory Cortex/pathology , Behavior, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Models, Animal , Neuroglia , Neurons/metabolism
5.
Commun Biol ; 3(1): 491, 2020 09 07.
Article in English | MEDLINE | ID: mdl-32895482

ABSTRACT

Whilst the brain is assumed to exert homeostatic functions to keep the cellular energy status constant under physiological conditions, this has not been experimentally proven. Here, we conducted in vivo optical recordings of intracellular concentration of adenosine 5'-triphosphate (ATP), the major cellular energy metabolite, using a genetically encoded sensor in the mouse brain. We demonstrate that intracellular ATP levels in cortical excitatory neurons fluctuate in a cortex-wide manner depending on the sleep-wake states, correlating with arousal. Interestingly, ATP levels profoundly decreased during rapid eye movement sleep, suggesting a negative energy balance in neurons despite a simultaneous increase in cerebral hemodynamics for energy supply. The reduction in intracellular ATP was also observed in response to local electrical stimulation for neuronal activation, whereas the hemodynamics were simultaneously enhanced. These observations indicate that cerebral energy metabolism may not always meet neuronal energy demands, consequently resulting in physiological fluctuations of intracellular ATP levels in neurons.


Subject(s)
Adenosine Triphosphate/metabolism , Cerebral Cortex/cytology , Intracellular Space/metabolism , Neurons/physiology , Sleep/physiology , Wakefulness/physiology , Animals , Cerebrovascular Circulation/physiology , Cortical Synchronization , Cytosol/metabolism , Electric Stimulation , Mice, Inbred C57BL , Optical Imaging
6.
Elife ; 82019 09 09.
Article in English | MEDLINE | ID: mdl-31496517

ABSTRACT

Hyperpolarization-activated cyclic-nucleotide-gated (HCN) channels control electrical rhythmicity and excitability in the heart and brain, but the function of HCN channels at the subcellular level in axons remains poorly understood. Here, we show that the action potential conduction velocity in both myelinated and unmyelinated central axons can be bidirectionally modulated by a HCN channel blocker, cyclic adenosine monophosphate (cAMP), and neuromodulators. Recordings from mouse cerebellar mossy fiber boutons show that HCN channels ensure reliable high-frequency firing and are strongly modulated by cAMP (EC50 40 µM; estimated endogenous cAMP concentration 13 µM). In addition, immunogold-electron microscopy revealed HCN2 as the dominating subunit in cerebellar mossy fibers. Computational modeling indicated that HCN2 channels control conduction velocity primarily by altering the resting membrane potential and are associated with significant metabolic costs. These results suggest that the cAMP-HCN pathway provides neuromodulators with an opportunity to finely tune energy consumption and temporal delays across axons in the brain.


Subject(s)
Action Potentials , Axons/physiology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Nerve Fibers/physiology , Neural Conduction , Potassium Channels/metabolism , Animals , Computer Simulation , Cyclic AMP/metabolism , Mice , Models, Neurological
8.
Acta Neuropathol ; 138(1): 147-161, 2019 07.
Article in English | MEDLINE | ID: mdl-30919030

ABSTRACT

Pelizaeus-Merzbacher disease (PMD) is an untreatable and fatal leukodystrophy. In a model of PMD with perturbed blood-brain barrier integrity, cholesterol supplementation promotes myelin membrane growth. Here, we show that in contrast to the mouse model, dietary cholesterol in two PMD patients did not lead to a major advancement of hypomyelination, potentially because the intact blood-brain barrier precludes its entry into the CNS. We therefore turned to a PMD mouse model with preserved blood-brain barrier integrity and show that a high-fat/low-carbohydrate ketogenic diet restored oligodendrocyte integrity and increased CNS myelination. This dietary intervention also ameliorated axonal degeneration and normalized motor functions. Moreover, in a paradigm of adult remyelination, ketogenic diet facilitated repair and attenuated axon damage. We suggest that a therapy with lipids such as ketone bodies, that readily enter the brain, can circumvent the requirement of a disrupted blood-brain barrier in the treatment of myelin disease.


Subject(s)
Demyelinating Diseases/pathology , Myelin Proteolipid Protein/metabolism , Oligodendroglia/physiology , Pelizaeus-Merzbacher Disease/pathology , Animals , Diet, Ketogenic , Disease Models, Animal , Mice , Oligodendroglia/metabolism , Organogenesis/physiology
9.
Elife ; 62017 04 17.
Article in English | MEDLINE | ID: mdl-28414271

ABSTRACT

In several neurodegenerative diseases and myelin disorders, the degeneration profiles of myelinated axons are compatible with underlying energy deficits. However, it is presently impossible to measure selectively axonal ATP levels in the electrically active nervous system. We combined transgenic expression of an ATP-sensor in neurons of mice with confocal FRET imaging and electrophysiological recordings of acutely isolated optic nerves. This allowed us to monitor dynamic changes and activity-dependent axonal ATP homeostasis at the cellular level and in real time. We find that changes in ATP levels correlate well with compound action potentials. However, this correlation is disrupted when metabolism of lactate is inhibited, suggesting that axonal glycolysis products are not sufficient to maintain mitochondrial energy metabolism of electrically active axons. The combined monitoring of cellular ATP and electrical activity is a novel tool to study neuronal and glial energy metabolism in normal physiology and in models of neurodegenerative disorders.


Subject(s)
Adenosine Triphosphate/analysis , Optic Nerve/chemistry , Optic Nerve/physiology , White Matter/chemistry , White Matter/physiology , Animals , Electroencephalography , Fluorescence Resonance Energy Transfer , Genes, Reporter , Mice , Mice, Transgenic , Microscopy, Confocal , Optical Imaging
10.
Neuron ; 91(1): 119-32, 2016 07 06.
Article in English | MEDLINE | ID: mdl-27292539

ABSTRACT

Oligodendrocytes make myelin and support axons metabolically with lactate. However, it is unknown how glucose utilization and glycolysis are adapted to the different axonal energy demands. Spiking axons release glutamate and oligodendrocytes express NMDA receptors of unknown function. Here we show that the stimulation of oligodendroglial NMDA receptors mobilizes glucose transporter GLUT1, leading to its incorporation into the myelin compartment in vivo. When myelinated optic nerves from conditional NMDA receptor mutants are challenged with transient oxygen-glucose deprivation, they show a reduced functional recovery when returned to oxygen-glucose but are indistinguishable from wild-type when provided with oxygen-lactate. Moreover, the functional integrity of isolated optic nerves, which are electrically silent, is extended by preincubation with NMDA, mimicking axonal activity, and shortened by NMDA receptor blockers. This reveals a novel aspect of neuronal energy metabolism in which activity-dependent glutamate release enhances oligodendroglial glucose uptake and glycolytic support of fast spiking axons.


Subject(s)
Axons/metabolism , Energy Metabolism/physiology , Glucose/metabolism , Oligodendroglia/metabolism , Optic Nerve/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Glucose Transporter Type 1/metabolism , Mice, Transgenic , Myelin Sheath/metabolism , Oxygen/metabolism
11.
Cell Metab ; 22(3): 361-3, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26331602

ABSTRACT

The discovery in mammals that axons are metabolically supported by myelinating glial cells explains why neurons can extend meters in length. In this issue, Volkenhoff et al. (2015) show that, in Drosophila, the transfer of lactate from the glial to the neuronal compartment is conserved in evolution, independent of body size.


Subject(s)
Blood-Brain Barrier/metabolism , Drosophila/physiology , Glycolysis , Neuroglia/metabolism , Neurons/metabolism , Trehalose/metabolism , Animals , Female
SELECTION OF CITATIONS
SEARCH DETAIL
...