Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.147
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 181(2): 396-409.e26, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32220308

ABSTRACT

Decades after the motor homunculus was first proposed, it is still unknown how different body parts are intermixed and interrelated in human motor cortical areas at single-neuron resolution. Using multi-unit recordings, we studied how face, head, arm, and leg movements are represented in the hand knob area of premotor cortex (precentral gyrus) in people with tetraplegia. Contrary to traditional expectations, we found strong representation of all movements and a partially "compositional" neural code that linked together all four limbs. The code consisted of (1) a limb-coding component representing the limb to be moved and (2) a movement-coding component where analogous movements from each limb (e.g., hand grasp and toe curl) were represented similarly. Compositional coding might facilitate skill transfer across limbs, and it provides a useful framework for thinking about how the motor system constructs movement. Finally, we leveraged these results to create a whole-body intracortical brain-computer interface that spreads targets across all limbs.


Subject(s)
Frontal Lobe/physiology , Motor Cortex/anatomy & histology , Motor Cortex/physiology , Adult , Brain Mapping , Frontal Lobe/anatomy & histology , Human Body , Humans , Motor Cortex/metabolism , Movement/physiology
2.
Cell ; 176(5): 1143-1157.e13, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30794775

ABSTRACT

We tested a newly described molecular memory system, CCR5 signaling, for its role in recovery after stroke and traumatic brain injury (TBI). CCR5 is uniquely expressed in cortical neurons after stroke. Post-stroke neuronal knockdown of CCR5 in pre-motor cortex leads to early recovery of motor control. Recovery is associated with preservation of dendritic spines, new patterns of cortical projections to contralateral pre-motor cortex, and upregulation of CREB and DLK signaling. Administration of a clinically utilized FDA-approved CCR5 antagonist, devised for HIV treatment, produces similar effects on motor recovery post stroke and cognitive decline post TBI. Finally, in a large clinical cohort of stroke patients, carriers for a naturally occurring loss-of-function mutation in CCR5 (CCR5-Δ32) exhibited greater recovery of neurological impairments and cognitive function. In summary, CCR5 is a translational target for neural repair in stroke and TBI and the first reported gene associated with enhanced recovery in human stroke.


Subject(s)
Brain Injuries, Traumatic/therapy , Receptors, CCR5/metabolism , Stroke/therapy , Aged , Aged, 80 and over , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Dendritic Spines/metabolism , Disease Models, Animal , Female , Humans , Male , Mice, Inbred C57BL , Middle Aged , Motor Cortex/metabolism , Neuronal Plasticity/physiology , Neurons/metabolism , Receptors, CCR5/physiology , Stroke Rehabilitation/methods
3.
Nature ; 625(7994): 345-351, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38057661

ABSTRACT

Frontotemporal lobar degeneration (FTLD) causes frontotemporal dementia (FTD), the most common form of dementia after Alzheimer's disease, and is often also associated with motor disorders1. The pathological hallmarks of FTLD are neuronal inclusions of specific, abnormally assembled proteins2. In the majority of cases the inclusions contain amyloid filament assemblies of TAR DNA-binding protein 43 (TDP-43) or tau, with distinct filament structures characterizing different FTLD subtypes3,4. The presence of amyloid filaments and their identities and structures in the remaining approximately 10% of FTLD cases are unknown but are widely believed to be composed of the protein fused in sarcoma (FUS, also known as translocated in liposarcoma). As such, these cases are commonly referred to as FTLD-FUS. Here we used cryogenic electron microscopy (cryo-EM) to determine the structures of amyloid filaments extracted from the prefrontal and temporal cortices of four individuals with FTLD-FUS. Surprisingly, we found abundant amyloid filaments of the FUS homologue TATA-binding protein-associated factor 15 (TAF15, also known as TATA-binding protein-associated factor 2N) rather than of FUS itself. The filament fold is formed from residues 7-99 in the low-complexity domain (LCD) of TAF15 and was identical between individuals. Furthermore, we found TAF15 filaments with the same fold in the motor cortex and brainstem of two of the individuals, both showing upper and lower motor neuron pathology. The formation of TAF15 amyloid filaments with a characteristic fold in FTLD establishes TAF15 proteinopathy in neurodegenerative disease. The structure of TAF15 amyloid filaments provides a basis for the development of model systems of neurodegenerative disease, as well as for the design of diagnostic and therapeutic tools targeting TAF15 proteinopathy.


Subject(s)
Frontotemporal Lobar Degeneration , TATA-Binding Protein Associated Factors , Humans , Amyloid/chemistry , Amyloid/metabolism , Amyloid/ultrastructure , Brain Stem/metabolism , Brain Stem/pathology , Cryoelectron Microscopy , Frontotemporal Dementia/etiology , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Frontotemporal Lobar Degeneration/complications , Frontotemporal Lobar Degeneration/metabolism , Frontotemporal Lobar Degeneration/pathology , Motor Cortex/metabolism , Motor Cortex/pathology , Motor Neurons/metabolism , Motor Neurons/pathology , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , TATA-Binding Protein Associated Factors/chemistry , TATA-Binding Protein Associated Factors/metabolism , TATA-Binding Protein Associated Factors/ultrastructure , Temporal Lobe/metabolism , Temporal Lobe/pathology
4.
Nature ; 624(7991): 390-402, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38092918

ABSTRACT

Divergence of cis-regulatory elements drives species-specific traits1, but how this manifests in the evolution of the neocortex at the molecular and cellular level remains unclear. Here we investigated the gene regulatory programs in the primary motor cortex of human, macaque, marmoset and mouse using single-cell multiomics assays, generating gene expression, chromatin accessibility, DNA methylome and chromosomal conformation profiles from a total of over 200,000 cells. From these data, we show evidence that divergence of transcription factor expression corresponds to species-specific epigenome landscapes. We find that conserved and divergent gene regulatory features are reflected in the evolution of the three-dimensional genome. Transposable elements contribute to nearly 80% of the human-specific candidate cis-regulatory elements in cortical cells. Through machine learning, we develop sequence-based predictors of candidate cis-regulatory elements in different species and demonstrate that the genomic regulatory syntax is highly preserved from rodents to primates. Finally, we show that epigenetic conservation combined with sequence similarity helps to uncover functional cis-regulatory elements and enhances our ability to interpret genetic variants contributing to neurological disease and traits.


Subject(s)
Conserved Sequence , Evolution, Molecular , Gene Expression Regulation , Gene Regulatory Networks , Mammals , Neocortex , Animals , Humans , Mice , Callithrix/genetics , Chromatin/genetics , Chromatin/metabolism , Conserved Sequence/genetics , DNA Methylation , DNA Transposable Elements/genetics , Epigenome , Gene Expression Regulation/genetics , Macaca/genetics , Mammals/genetics , Motor Cortex/cytology , Motor Cortex/metabolism , Multiomics , Neocortex/cytology , Neocortex/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Single-Cell Analysis , Transcription Factors/metabolism , Genetic Variation/genetics
5.
Nature ; 601(7891): 139-143, 2022 01.
Article in English | MEDLINE | ID: mdl-34880495

ABSTRACT

The abnormal aggregation of TAR DNA-binding protein 43 kDa (TDP-43) in neurons and glia is the defining pathological hallmark of the neurodegenerative disease amyotrophic lateral sclerosis (ALS) and multiple forms of frontotemporal lobar degeneration (FTLD)1,2. It is also common in other diseases, including Alzheimer's and Parkinson's. No disease-modifying therapies exist for these conditions and early diagnosis is not possible. The structures of pathological TDP-43 aggregates are unknown. Here we used cryo-electron microscopy to determine the structures of aggregated TDP-43 in the frontal and motor cortices of an individual who had ALS with FTLD and from the frontal cortex of a second individual with the same diagnosis. An identical amyloid-like filament structure comprising a single protofilament was found in both brain regions and individuals. The ordered filament core spans residues 282-360 in the TDP-43 low-complexity domain and adopts a previously undescribed double-spiral-shaped fold, which shows no similarity to those of TDP-43 filaments formed in vitro3,4. An abundance of glycine and neutral polar residues facilitates numerous turns and restricts ß-strand length, which results in an absence of ß-sheet stacking that is associated with cross-ß amyloid structure. An uneven distribution of residues gives rise to structurally and chemically distinct surfaces that face external densities and suggest possible ligand-binding sites. This work enhances our understanding of the molecular pathogenesis of ALS and FTLD and informs the development of diagnostic and therapeutic agents that target aggregated TDP-43.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Cryoelectron Microscopy , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/ultrastructure , Frontotemporal Lobar Degeneration/metabolism , Frontotemporal Lobar Degeneration/pathology , Amino Acid Sequence , Amyloid beta-Peptides/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Frontal Lobe/metabolism , Frontal Lobe/pathology , Frontal Lobe/ultrastructure , Humans , Male , Middle Aged , Motor Cortex/metabolism , Motor Cortex/pathology , Motor Cortex/ultrastructure , Mutation
6.
Brain ; 147(8): 2826-2841, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38643019

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a severe motor neuron disease with uncertain genetic predisposition in most sporadic cases. The spatial architecture of cell types and gene expression are the basis of cell-cell interactions, biological function and disease pathology, but are not well investigated in the human motor cortex, a key ALS-relevant brain region. Recent studies indicated single nucleus transcriptomic features of motor neuron vulnerability in ALS motor cortex. However, the brain regional vulnerability of ALS-associated genes and the genetic link between region-specific genes and ALS risk remain largely unclear. Here, we developed an entropy-weighted differential gene expression matrix-based tool (SpatialE) to identify the spatial enrichment of gene sets in spatial transcriptomics. We benchmarked SpatialE against another enrichment tool (multimodal intersection analysis) using spatial transcriptomics data from both human and mouse brain tissues. To investigate regional vulnerability, we analysed three human motor cortex and two dorsolateral prefrontal cortex tissues for spatial enrichment of ALS-associated genes. We also used Cell2location to estimate the abundance of cell types in ALS-related cortex layers. To dissect the link of regionally expressed genes and ALS risk, we performed burden analyses of rare loss-of-function variants detected by whole-genome sequencing in ALS patients and controls, then analysed differential gene expression in the TargetALS RNA-sequencing dataset. SpatialE showed more accurate and specific spatial enrichment of regional cell type markers than multimodal intersection analysis in both mouse brain and human dorsolateral prefrontal cortex. Spatial transcriptomic analyses of human motor cortex showed heterogeneous cell types and spatial gene expression profiles. We found that 260 manually curated ALS-associated genes are significantly enriched in layer 5 of the motor cortex, with abundant expression of upper motor neurons and layer 5 excitatory neurons. Burden analyses of rare loss-of-function variants in Layer 5-associated genes nominated NOMO1 as a novel ALS-associated gene in a combined sample set of 6814 ALS patients and 3324 controls (P = 0.029). Gene expression analyses in CNS tissues revealed downregulation of NOMO1 in ALS, which is consistent with a loss-of-function disease mechanism. In conclusion, our integrated spatial transcriptomics and genomic analyses identified regional brain vulnerability in ALS and the association of a layer 5 gene (NOMO1) with ALS risk.


Subject(s)
Amyotrophic Lateral Sclerosis , Motor Cortex , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Humans , Mice , Animals , Motor Cortex/metabolism , Motor Cortex/pathology , Transcriptome , Genomics/methods , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Motor Neurons/metabolism , Motor Neurons/pathology , Male
7.
Cell Mol Life Sci ; 81(1): 346, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39134808

ABSTRACT

In people with multiple sclerosis (MS), newborn and surviving oligodendrocytes (OLs) can contribute to remyelination, however, current therapies are unable to enhance or sustain endogenous repair. Low intensity repetitive transcranial magnetic stimulation (LI-rTMS), delivered as an intermittent theta burst stimulation (iTBS), increases the survival and maturation of newborn OLs in the healthy adult mouse cortex, but it is unclear whether LI-rTMS can promote remyelination. To examine this possibility, we fluorescently labelled oligodendrocyte progenitor cells (OPCs; Pdgfrα-CreER transgenic mice) or mature OLs (Plp-CreER transgenic mice) in the adult mouse brain and traced the fate of each cell population over time. Daily sessions of iTBS (600 pulses; 120 mT), delivered during cuprizone (CPZ) feeding, did not alter new or pre-existing OL survival but increased the number of myelin internodes elaborated by new OLs in the primary motor cortex (M1). This resulted in each new M1 OL producing ~ 471 µm more myelin. When LI-rTMS was delivered after CPZ withdrawal (during remyelination), it significantly increased the length of the internodes elaborated by new M1 and callosal OLs, increased the number of surviving OLs that supported internodes in the corpus callosum (CC), and increased the proportion of axons that were myelinated. The ability of LI-rTMS to modify cortical neuronal activity and the behaviour of new and surviving OLs, suggests that it may be a suitable adjunct intervention to enhance remyelination in people with MS.


Subject(s)
Cuprizone , Demyelinating Diseases , Myelin Sheath , Oligodendroglia , Remyelination , Transcranial Magnetic Stimulation , Animals , Transcranial Magnetic Stimulation/methods , Oligodendroglia/metabolism , Demyelinating Diseases/therapy , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Mice , Myelin Sheath/metabolism , Disease Models, Animal , Mice, Transgenic , Motor Cortex/pathology , Motor Cortex/metabolism , Cell Survival , Mice, Inbred C57BL , Multiple Sclerosis/therapy , Multiple Sclerosis/pathology
8.
J Neurosci ; 43(15): 2696-2713, 2023 04 12.
Article in English | MEDLINE | ID: mdl-36894315

ABSTRACT

Although motor cortex is crucial for learning precise and reliable movements, whether and how astrocytes contribute to its plasticity and function during motor learning is unknown. Here, we report that astrocyte-specific manipulations in primary motor cortex (M1) during a lever push task alter motor learning and execution, as well as the underlying neuronal population coding. Mice that express decreased levels of the astrocyte glutamate transporter 1 (GLT1) show impaired and variable movement trajectories, whereas mice with increased astrocyte Gq signaling show decreased performance rates, delayed response times, and impaired trajectories. In both groups, which include male and female mice, M1 neurons have altered interneuronal correlations and impaired population representations of task parameters, including response time and movement trajectories. RNA sequencing further supports a role for M1 astrocytes in motor learning and shows changes in astrocytic expression of glutamate transporter genes, GABA transporter genes, and extracellular matrix protein genes in mice that have acquired this learned behavior. Thus, astrocytes coordinate M1 neuronal activity during motor learning, and our results suggest that this contributes to learned movement execution and dexterity through mechanisms that include regulation of neurotransmitter transport and calcium signaling.SIGNIFICANCE STATEMENT We demonstrate for the first time that in the M1 of mice, astrocyte function is critical for coordinating neuronal population activity during motor learning. We demonstrate that knockdown of astrocyte glutamate transporter GLT1 affects specific components of learning, such as smooth trajectory formation. Altering astrocyte calcium signaling by activation of Gq-DREADD upregulates GLT1 and affects other components of learning, such as response rates and reaction times as well as trajectory smoothness. In both manipulations, neuronal activity in motor cortex is dysregulated, but in different ways. Thus, astrocytes have a crucial role in motor learning via their influence on motor cortex neurons, and they do so by mechanisms that include regulation of glutamate transport and calcium signals.


Subject(s)
Astrocytes , Motor Cortex , Mice , Male , Animals , Female , Astrocytes/metabolism , Motor Cortex/metabolism , Motor Neurons/metabolism , Synaptic Transmission , Amino Acid Transport System X-AG/metabolism
9.
Eur J Neurosci ; 59(2): 177-191, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38049944

ABSTRACT

Microglia are essential contributors to synaptic transmission and stability and communicate with neurons via the fractalkine pathway. Transcranial direct current stimulation [(t)DCS], a form of non-invasive electrical brain stimulation, modulates cortical excitability and promotes neuroplasticity, which has been extensively demonstrated in the motor cortex and for motor learning. The role of microglia and their fractalkine receptor CX3CR1 in motor cortical neuroplasticity mediated by DCS or motor learning requires further elucidation. We demonstrate the effects of pharmacological microglial depletion and genetic Cx3cr1 deficiency on the induction of DCS-induced long-term potentiation (DCS-LTP) ex vivo. The relevance of microglia-neuron communication for DCS response and structural neuroplasticity underlying motor learning are assessed via 2-photon in vivo imaging. The behavioural consequences of impaired CX3CR1 signalling are investigated for both gross and fine motor learning. We show that DCS-mediated neuroplasticity in the motor cortex depends on the presence of microglia and is driven in part by CX3CR1 signalling ex vivo and provide the first evidence of microglia interacting with neurons during DCS in vivo. Furthermore, CX3CR1 signalling is required for motor learning and underlying structural neuroplasticity in concert with microglia interaction. Although we have recently demonstrated the microglial response to DCS in vivo, we now provide a link between microglial integrity and neuronal activity for the expression of DCS-dependent neuroplasticity. In addition, we extend the knowledge on the relevance of CX3CR1 signalling for motor learning and structural neuroplasticity. The underlying molecular mechanisms and the potential impact of DCS in rescuing CX3CR1 deficits remain to be addressed in the future.


Subject(s)
Motor Cortex , Transcranial Direct Current Stimulation , Motor Cortex/metabolism , Neurons/metabolism , Microglia/metabolism , Neuronal Plasticity/physiology , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism
10.
Acta Neuropathol ; 147(1): 100, 2024 06 17.
Article in English | MEDLINE | ID: mdl-38884646

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a rapidly progressive neurodegenerative disease with average lifespan of 2-5 years after diagnosis. The identification of novel prognostic and pharmacodynamic biomarkers are needed to facilitate therapeutic development. Metalloprotein human superoxide dismutase 1 (SOD1) is known to accumulate and form aggregates in patient neural tissue with familial ALS linked to mutations in their SOD1 gene. Aggregates of SOD1 have also been detected in other forms of ALS, including the sporadic form and the most common familial form linked to abnormal hexanucleotide repeat expansions in the Chromosome 9 open reading frame 72 (C9ORF72) gene. Here, we report the development of a real-time quaking-induced conversion (RT-QuIC) seed amplification assay using a recombinant human SOD1 substrate to measure SOD1 seeding activity in postmortem spinal cord and motor cortex tissue from persons with different ALS etiologies. Our SOD1 RT-QuIC assay detected SOD1 seeds in motor cortex and spinal cord dilutions down to 10-5. Importantly, we detected SOD1 seeding activity in specimens from both sporadic and familial ALS cases, with the latter having mutations in either their SOD1 or C9ORF72 genes. Analyses of RT-QuIC parameters indicated similar lag phases in spinal cords of sporadic and familial ALS patients, but higher ThT fluorescence maxima by SOD1 familial ALS specimens and sporadic ALS thoracic cord specimens. For a subset of sporadic ALS patients, motor cortex and spinal cords were examined, with seeding activity in both anatomical regions. Our results suggest SOD1 seeds are in ALS patient neural tissues not linked to SOD1 mutation, suggesting that SOD1 seeding activity may be a promising biomarker, particularly in sporadic ALS cases for whom genetic testing is uninformative.


Subject(s)
Amyotrophic Lateral Sclerosis , Biomarkers , Spinal Cord , Superoxide Dismutase-1 , Aged , Female , Humans , Male , Middle Aged , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/metabolism , C9orf72 Protein/genetics , Motor Cortex/pathology , Motor Cortex/metabolism , Mutation/genetics , Spinal Cord/pathology , Spinal Cord/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Biomarkers/analysis
11.
Brain Behav Immun ; 119: 693-708, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38677626

ABSTRACT

Newborns exposed to birth asphyxia transiently experience deficient blood flow and a lack of oxygen, potentially inducing hypoxic-ischaemic encephalopathy and subsequent neurological damage. Immunomodulatory components in plasma may dampen these responses. Using caesarean-delivered pigs as a model, we hypothesized that dietary plasma supplementation improves brain outcomes in pigs exposed to birth asphyxia. Mild birth asphyxia was induced by temporary occlusion of the umbilical cord prior to caesarean delivery. Motor development was assessed in asphyxiated (ASP) and control (CON) piglets using neonatal arousal, physical activity and gait test parameters before euthanasia on Day 4. The ASP pigs exhibited increased plasma lactate at birth, deficient motor skills and increased glial fibrillary acidic protein levels in CSF and astrogliosis in the putamen. The expression of genes related to oxidative stress, inflammation and synaptic functions was transiently altered in the motor cortex and caudate nucleus. The number of apoptotic cells among CTIP2-positive neurons in the motor cortex and striatal medium spiny neurons was increased, and maturation of preoligodendrocytes in the internal capsule was delayed. Plasma supplementation improved gait performance in the beam test, attenuated neuronal apoptosis and affected gene expression related to neuroinflammation, neurotransmission and antioxidants (motor cortex, caudate). We present a new clinically relevant animal model of moderate birth asphyxia inducing structural and functional brain damage. The components in plasma that support brain repair remain to be identified but may represent a therapeutic potential for infants and animals after birth asphyxia.


Subject(s)
Animals, Newborn , Asphyxia Neonatorum , Brain , Disease Models, Animal , Animals , Swine , Asphyxia Neonatorum/therapy , Brain/metabolism , Female , Oxidative Stress/physiology , Hypoxia-Ischemia, Brain/therapy , Hypoxia-Ischemia, Brain/metabolism , Neurons/metabolism , Glial Fibrillary Acidic Protein/metabolism , Asphyxia/therapy , Pregnancy , Motor Cortex/metabolism
12.
Cell Mol Biol (Noisy-le-grand) ; 70(4): 53-60, 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38678627

ABSTRACT

Cobalt protoporphyrin (CoPP) is a synthetic heme analog that has been observed to reduce food intake and promote sustained weight loss. While the precise mechanisms responsible for these effects remain elusive, earlier research has hinted at the potential involvement of nitric oxide synthase in the hypothalamus. This study aimed to delve into CoPP's impact on the activities of crucial antioxidant enzymes: superoxide dismutase (SOD), catalase, glutathione peroxidase (GPx), glutathione reductase (GR), and glutathione-S-transferase (GST) across seven distinct brain regions (hippocampus, hypothalamus, prefrontal cortex, motor cortex, striatum, midbrain, and cerebellum), as well as in the liver and kidneys. Female Wistar rats weighing 180 to 200 grams received a single subcutaneous dose of 25 µmol/kg CoPP. After six days, brain tissue was extracted to assess the activities of antioxidant enzymes and quantify malondialdehyde levels. Our findings confirm that CoPP administration triggers the characteristic effects of decreased food intake and reduced body weight. Moreover, it led to an increase in SOD activity in the hypothalamus, a pivotal brain region associated with food intake regulation. Notably, CoPP-treated rats exhibited elevated enzymatic activity of catalase, GR, and GST in the motor cortex without concurrent signs of heightened oxidative stress. These results underscore a strong connection between the antioxidant system and food intake regulation. They also emphasize the need for further investigation into the roles of antioxidant enzymes in modulating food intake and the ensuing weight loss, using CoPP as a valuable research tool.


Subject(s)
Antioxidants , Hypothalamus , Motor Cortex , Protoporphyrins , Animals , Female , Rats , Antioxidants/metabolism , Body Weight/drug effects , Catalase/metabolism , Eating/drug effects , Glutathione Peroxidase/drug effects , Glutathione Peroxidase/metabolism , Glutathione Reductase/drug effects , Glutathione Reductase/metabolism , Glutathione Transferase/drug effects , Glutathione Transferase/metabolism , Hypothalamus/metabolism , Hypothalamus/drug effects , Hypothalamus/enzymology , Malondialdehyde/metabolism , Motor Cortex/drug effects , Motor Cortex/metabolism , Motor Cortex/enzymology , Oxidative Stress/drug effects , Protoporphyrins/pharmacology , Rats, Wistar , Superoxide Dismutase/drug effects , Superoxide Dismutase/metabolism
13.
Nature ; 563(7729): 72-78, 2018 11.
Article in English | MEDLINE | ID: mdl-30382198

ABSTRACT

The neocortex contains a multitude of cell types that are segregated into layers and functionally distinct areas. To investigate the diversity of cell types across the mouse neocortex, here we analysed 23,822 cells from two areas at distant poles of the mouse neocortex: the primary visual cortex and the anterior lateral motor cortex. We define 133 transcriptomic cell types by deep, single-cell RNA sequencing. Nearly all types of GABA (γ-aminobutyric acid)-containing neurons are shared across both areas, whereas most types of glutamatergic neurons were found in one of the two areas. By combining single-cell RNA sequencing and retrograde labelling, we match transcriptomic types of glutamatergic neurons to their long-range projection specificity. Our study establishes a combined transcriptomic and projectional taxonomy of cortical cell types from functionally distinct areas of the adult mouse cortex.


Subject(s)
Gene Expression Profiling , Neocortex/cytology , Neocortex/metabolism , Animals , Biomarkers/analysis , Female , GABAergic Neurons/metabolism , Glutamic Acid/metabolism , Male , Mice , Motor Cortex/anatomy & histology , Motor Cortex/cytology , Motor Cortex/metabolism , Neocortex/anatomy & histology , Organ Specificity , Sequence Analysis, RNA , Single-Cell Analysis , Visual Cortex/anatomy & histology , Visual Cortex/cytology , Visual Cortex/metabolism
14.
Mol Cell Neurosci ; 126: 103883, 2023 09.
Article in English | MEDLINE | ID: mdl-37527694

ABSTRACT

There is growing interest in the use of natural products for the treatment of Parkinson's disease (PD). Mucuna pruriens has been used in the treatment of humans with PD. The goal of this study was to determine if daily oral treatment with an extract of Mucuna pruriens, starting after the MPTP-induced loss of nigrostriatal dopamine in male mice, would result in recovery/restoration of motor function, tyrosine hydroxylase (TH) protein expression in the nigrostriatal pathway, or glutamate biomarkers in both the striatum and motor cortex. Following MPTP administration, resulting in an 80 % loss of striatal TH, treatment with Mucuna pruriens failed to rescue either striatal TH or the dopamine transporter back to the control levels, but there was restoration of gait/motor function. There was an MPTP-induced loss of TH-labeled neurons in the substantia nigra pars compacta and in the number of striatal dendritic spines, both of which failed to be recovered following treatment with Mucuna pruriens. This Mucuna pruriens-induced locomotor recovery following MPTP was associated with restoration of two striatal glutamate transporter proteins, GLAST (EAAT1) and EAAC1 (EAAT3), and the vesicular glutamate transporter 2 (Vglut2) within the motor cortex. Post-MPTP treatment with Mucuna pruriens, results in locomotor improvement that is associated with recovery of striatal and motor cortex glutamate transporters but is independent of nigrostriatal TH restoration.


Subject(s)
Mucuna , Parkinson Disease , Plant Extracts , Parkinson Disease/drug therapy , Parkinson Disease/pathology , Glutamic Acid/metabolism , Biomarkers/metabolism , Motor Cortex/drug effects , Motor Cortex/metabolism , Motor Cortex/pathology , Mucuna/chemistry , Plant Extracts/administration & dosage , Gait/drug effects , Pars Compacta/metabolism , Pars Compacta/pathology , Basal Ganglia/metabolism , Basal Ganglia/pathology , Animals , Mice
15.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Article in English | MEDLINE | ID: mdl-33443203

ABSTRACT

Activity of sensory and motor cortices is essential for sensorimotor integration. In particular, coherence between these areas may indicate binding of critical functions like perception, motor planning, action, or sleep. Evidence is accumulating that cerebellar output modulates cortical activity and coherence, but how, when, and where it does so is unclear. We studied activity in and coherence between S1 and M1 cortices during whisker stimulation in the absence and presence of optogenetic Purkinje cell stimulation in crus 1 and 2 of awake mice, eliciting strong simple spike rate modulation. Without Purkinje cell stimulation, whisker stimulation triggers fast responses in S1 and M1 involving transient coherence in a broad spectrum. Simultaneous stimulation of Purkinje cells and whiskers affects amplitude and kinetics of sensory responses in S1 and M1 and alters the estimated S1-M1 coherence in theta and gamma bands, allowing bidirectional control dependent on behavioral context. These effects are absent when Purkinje cell activation is delayed by 20 ms. Focal stimulation of Purkinje cells revealed site specificity, with cells in medial crus 2 showing the most prominent and selective impact on estimated coherence, i.e., a strong suppression in the gamma but not the theta band. Granger causality analyses and computational modeling of the involved networks suggest that Purkinje cells control S1-M1 phase consistency predominantly via ventrolateral thalamus and M1. Our results indicate that activity of sensorimotor cortices can be dynamically and functionally modulated by specific cerebellar inputs, highlighting a widespread role of the cerebellum in coordinating sensorimotor behavior.


Subject(s)
Motor Cortex/metabolism , Purkinje Cells/metabolism , Somatosensory Cortex/metabolism , Animals , Cerebellar Cortex , Cerebellum/metabolism , Female , Humans , Male , Mice , Mice, Transgenic , Optogenetics , Sensorimotor Cortex , Ventral Thalamic Nuclei , Vibrissae/physiology
16.
Int J Mol Sci ; 25(17)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39273491

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder characterized by motor symptoms like tremors and bradykinesia. PD's pathology involves the aggregation of α-synuclein and loss of dopaminergic neurons, leading to altered neural oscillations in the cortico-basal ganglia-thalamic network. Despite extensive research, the relationship between the motor symptoms of PD and transient changes in brain oscillations before and after motor tasks in different brain regions remain unclear. This study aimed to investigate neural oscillations in both healthy and PD model mice using local field potential (LFP) recordings from multiple brain regions during rest and locomotion. The histological evaluation confirmed the significant dopaminergic neuron loss in the injection side in 6-OHDA lesioned mice. Behavioral tests showed motor deficits in these mice, including impaired coordination and increased forelimb asymmetry. The LFP analysis revealed increased delta, theta, alpha, beta, and gamma band activity in 6-OHDA lesioned mice during movement, with significant increases in multiple brain regions, including the primary motor cortex (M1), caudate-putamen (CPu), subthalamic nucleus (STN), substantia nigra pars compacta (SNc), and pedunculopontine nucleus (PPN). Taken together, these results show that the motor symptoms of PD are accompanied by significant transient increases in brain oscillations, especially in the gamma band. This study provides potential biomarkers for early diagnosis and therapeutic evaluation by elucidating the relationship between specific neural oscillations and motor deficits in PD.


Subject(s)
Disease Models, Animal , Parkinson Disease , Animals , Mice , Parkinson Disease/physiopathology , Male , Oxidopamine , Mice, Inbred C57BL , Motor Cortex/physiopathology , Motor Cortex/metabolism , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Brain/physiopathology , Brain/pathology , Brain/metabolism , Brain Waves , Motor Activity
17.
Int J Mol Sci ; 25(16)2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39201305

ABSTRACT

Compensatory changes in brain connectivity keep motor symptoms mild in prodromal Parkinson's disease. Studying compensation in patients is hampered by the steady progression of the disease and a lack of individual baseline controls. Furthermore, combining fMRI with walking is intricate. We therefore used a seed-based metabolic connectivity analysis based on 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) uptake in a unilateral 6-OHDA rat model. At baseline and in the chronic phase 6-7 months after lesion, rats received an intraperitoneal injection of [18F]FDG and spent 50 min walking on a horizontal treadmill, followed by a brain PET-scan under anesthesia. High activity was found in the cerebellar anterior vermis in both conditions. At baseline, the anterior vermis showed hardly any stable connections to the rest of the brain. The (future) ipsilesional cerebellar hemisphere was not particularly active during walking but was extensively connected to many brain areas. After unilateral dopamine depletion, rats still walked normally without obvious impairments. The ipsilesional cerebellar hemisphere increased its activity, but narrowed its connections down to the vestibulocerebellum, probably aiding lateral stability. The anterior vermis established a network involving the motor cortex, hippocampus and thalamus. Adding those regions to the vermis network of (previously) automatic control of locomotion suggests that after unilateral dopamine depletion considerable conscious and cognitive effort has to be provided to achieve stable walking.


Subject(s)
Cerebellum , Dopamine , Positron-Emission Tomography , Walking , Animals , Rats , Dopamine/metabolism , Walking/physiology , Cerebellum/metabolism , Cerebellum/diagnostic imaging , Male , Fluorodeoxyglucose F18/metabolism , Magnetic Resonance Imaging/methods , Oxidopamine , Exercise Test , Disease Models, Animal , Motor Cortex/metabolism , Motor Cortex/diagnostic imaging , Motor Cortex/physiopathology
18.
J Neurosci ; 42(43): 8095-8112, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36104281

ABSTRACT

Intracortical inhibition in motor cortex (M1) regulates movement and motor learning. If cortical and thalamic inputs target different inhibitory cell types in different layers, then these afferents may play different roles in regulating M1 output. Using mice of both sexes, we quantified input to two main classes of M1 interneurons, parvalbumin+ (PV+) cells and somatostatin+ (SOM+) cells, using monosynaptic rabies tracing. We then compared anatomic and functional connectivity based on synaptic strength from sensory cortex and thalamus. Functionally, each input innervated M1 interneurons with a unique laminar profile. Different interneuron types were excited in a distinct, complementary manner, suggesting feedforward inhibition proceeds selectively via distinct circuits. Specifically, somatosensory cortex (S1) inputs primarily targeted PV+ neurons in upper layers (L2/3) but SOM+ neurons in middle layers (L5). Somatosensory thalamus [posterior nucleus (PO)] inputs targeted PV+ neurons in middle layers (L5). In contrast to sensory cortical areas, thalamic input to SOM+ neurons was equivalent to that of PV+ neurons. Thus, long-range excitatory inputs target inhibitory neurons in an area and a cell type-specific manner, which contrasts with input to neighboring pyramidal cells. In contrast to feedforward inhibition providing generic inhibitory tone in cortex, circuits are selectively organized to recruit inhibition matched to incoming excitatory circuits.SIGNIFICANCE STATEMENT M1 integrates sensory information and frontal cortical inputs to plan and control movements. Although inputs to excitatory cells are described, the synaptic circuits by which these inputs drive specific types of M1 interneurons are unknown. Anatomical results with rabies tracing and physiological quantification of synaptic strength shows that two main classes of inhibitory cells (PV+ and SOM+ interneurons) both receive substantial cortical and thalamic input, in contrast to interneurons in sensory areas (where thalamic input strongly prefers PV+ interneurons). Further, each input studied targets PV+ and SOM+ interneurons in a different fashion, suggesting that separate, specific circuits exist for recruitment of feedforward inhibition.


Subject(s)
Motor Cortex , Rabies , Female , Male , Mice , Animals , Parvalbumins/metabolism , Motor Cortex/metabolism , Rabies/metabolism , Thalamus/physiology , Neurons/physiology , Interneurons/physiology , Somatostatin/metabolism
19.
Neuroimage ; 284: 120465, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37993003

ABSTRACT

Neural-activity-associated hemodynamic changes have been used to noninvasively measure brain function in the early developmental stages. However, the temporal changes in their hemodynamics are not always consistent with adults. Studies have not evaluated developmental changes for a long period using the same stimuli; therefore, this study examined the normalized relative changes in oxygenated hemoglobin (Δ[oxy-Hb]) in full-term infants and compared them with neonates up to 10 months of age during the administration of tactile vibration stimuli to their limbs using whole-head functional near-infrared spectroscopy. The time to peak of normalized Δ[oxy-Hb] was not affected by age. The amplitude of normalized Δ[oxy-Hb] showed an effect of age in broader areas, including sensorimotor-related but excluding supplementary motor area; the amplitude of normalized Δ[oxy-Hb] decreased the most in the 1-2-month-old group and later increased with development. We hypothesized that these results may reflect developmental changes in neural activity, vasculature, and blood oxygenation.


Subject(s)
Motor Cortex , Spectroscopy, Near-Infrared , Adult , Infant, Newborn , Infant , Humans , Spectroscopy, Near-Infrared/methods , Hemodynamics/physiology , Oxyhemoglobins/analysis , Oxyhemoglobins/metabolism , Motor Cortex/metabolism , Touch , Hemoglobins/metabolism
20.
Neuroimage ; 266: 119830, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36566925

ABSTRACT

Aging is associated with alterations in the brain including structural and metabolic changes. Previous research has focused on neurometabolite level differences associated to age in a variety of brain regions, but the relationship among metabolites across the brain has been much less studied. Investigating these relationships can reveal underlying neurometabolic processes, their interdependency, and their progress throughout the lifespan. Using 1H-MRS, we investigated the relationship among metabolite concentrations of N-acetylaspartate (NAA), creatine (Cr), choline (Cho), myo-Inositol (mIns) and glutamate-glutamine complex (Glx) in seven voxel locations, i.e., bilateral sensorimotor cortex, bilateral striatum, pre-supplementary motor area, right inferior frontal gyrus and occipital cortex. These measurements were performed on 59 human participants divided in two age groups: young adults (YA: 23.2 ± 4.3; 18-34 years) and older adults (OA: 67.5 ± 3.9; 61-74 years). Our results showed age-related differences in NAA, Cho, and mIns across brain regions, suggesting the presence of neurodegeneration and altered gliosis. Moreover, associative patterns among NAA, Cho and Cr were observed across the selected brain regions, which differed between young and older adults. Whereas most of metabolite concentrations were inhomogeneous across different brain regions, Cho levels were shown to be strongly related across brain regions in both age groups. Finally, we found metabolic associations between homologous brain regions (SM1 and striatum) in the OA group, with NAA showing a significant correlation between bilateral sensorimotor cortices (SM1) and mIns levels being correlated between the bilateral striata. We posit that a network perspective provides important insights regarding the potential interactions among neurochemicals underlying metabolic processes at a local and global level and their relationship with aging.


Subject(s)
Motor Cortex , Sensorimotor Cortex , Young Adult , Humans , Aged , Proton Magnetic Resonance Spectroscopy , Brain/diagnostic imaging , Brain/metabolism , Aging , Motor Cortex/metabolism , Sensorimotor Cortex/metabolism , Prefrontal Cortex/metabolism , Aspartic Acid , Creatine/metabolism , Choline/metabolism , Inositol/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL