Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.533
Filter
1.
J Adv Res ; 56: 125-136, 2024 Feb.
Article in English | MEDLINE | ID: mdl-36940850

ABSTRACT

INTRODUCTION: The glymphatic system offers a perivascular pathway for the clearance of pathological proteins and metabolites to optimize neurological functions. Glymphatic dysfunction plays a pathogenic role in Parkinson's disease (PD); however, the molecular mechanism of glymphatic dysfunction in PD remains elusive. OBJECTIVE: To explore whether matrix metalloproteinase-9 (MMP-9)-mediated ß-dystroglycan (ß-DG) cleavage is involved in the regulation of aquaporin-4 (AQP4) polarity-mediated glymphatic system in PD. METHODS: 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD and A53T mice were used in this study. The glymphatic function was evaluated using ex vivo imaging. TGN-020, an AQP4 antagonist, was administered to investigate the role of AQP4 in glymphatic dysfunction in PD. GM6001, an MMP-9 antagonist, was administered to investigate the role of the MMP-9/ß-DG pathway in regulating AQP4. The expression and distribution of AQP4, MMP-9, and ß-DG were assessed using western blotting, immunofluorescence, and co-immunoprecipitation. The ultrastructure of basement membrane (BM)-astrocyte endfeet was detected using transmission electron microscopy. Rotarod and open-field tests were performed to evaluate motor behavior. RESULTS: Perivascular influx and efflux of cerebral spinal fluid tracers were reduced in MPTP-induced PD mice with impaired AQP4 polarization. AQP4 inhibition aggravated reactive astrogliosis, glymphatic drainage restriction, and dopaminergic neuronal loss in MPTP-induced PD mice. MMP-9 and cleaved ß-DG were upregulated in both MPTP-induced PD and A53T mice, with reduced polarized localization of ß-DG and AQP4 to astrocyte endfeet. MMP-9 inhibition restored BM-astrocyte endfeet-AQP4 integrity and attenuated MPTP-induced metabolic perturbations and dopaminergic neuronal loss. CONCLUSION: AQP4 depolarization contributes to glymphatic dysfunction and aggravates PD pathologies, and MMP-9-mediated ß-DG cleavage regulates glymphatic function through AQP4 polarization in PD, which may provide novel insights into the pathogenesis of PD.


Subject(s)
Aquaporins , Glymphatic System , Parkinson Disease , Mice , Animals , Parkinson Disease/metabolism , Parkinson Disease/pathology , Astrocytes/metabolism , Astrocytes/pathology , Astrocytes/ultrastructure , Matrix Metalloproteinase 9/metabolism , Glymphatic System/metabolism , Dopamine/metabolism , Aquaporins/metabolism
2.
Science ; 378(6619): eadc9020, 2022 11 04.
Article in English | MEDLINE | ID: mdl-36378959

ABSTRACT

Astrocytes, a type of glia, are abundant and morphologically complex cells. Here, we report astrocyte molecular profiles, diversity, and morphology across the mouse central nervous system (CNS). We identified shared and region-specific astrocytic genes and functions and explored the cellular origins of their regional diversity. We identified gene networks correlated with astrocyte morphology, several of which unexpectedly contained Alzheimer's disease (AD) risk genes. CRISPR/Cas9-mediated reduction of candidate genes reduced astrocyte morphological complexity and resulted in cognitive deficits. The same genes were down-regulated in human AD, in an AD mouse model that displayed reduced astrocyte morphology, and in other human brain disorders. We thus provide comprehensive molecular data on astrocyte diversity and mechanisms across the CNS and on the molecular basis of astrocyte morphology in health and disease.


Subject(s)
Alzheimer Disease , Astrocytes , Central Nervous System , Transcriptome , Animals , Humans , Mice , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Astrocytes/classification , Astrocytes/metabolism , Astrocytes/ultrastructure , Disease Models, Animal , Central Nervous System/cytology , Central Nervous System/metabolism
3.
Sci Rep ; 12(1): 1685, 2022 01 31.
Article in English | MEDLINE | ID: mdl-35102230

ABSTRACT

Repeat-associated non-AUG (RAN) translation of mRNAs/transcripts responsible for polyglutamine (polyQ) diseases may generate peptides containing different mono amino acid tracts such as polyserine (polyS) and polyleucine (polyL). The propagation of aggregated polyQ from one cell to another is also an intriguing feature of polyQ proteins. However, whether the RAN translation-related polyS and polyL have the ability to propagate remains unclear, and if they do, whether the exogenous polyS and polyL exert toxicity on the recipient cells is also not known yet. In the present study, we found that aggregated polyS and polyL peptides spontaneously enter neuron-like cells and astrocytes in vitro. Aggregated polyS led to the degeneration of the differentiated neuron-like cultured cells. Likewise, the two types of aggregates taken up by astrocytes induced aberrant differentiation and cell death in vitro. Furthermore, injection of each of the two types of aggregates into the ventricles of adult mice resulted in their behavioral changes. The polyS-injected mice showed extensive vacuolar degeneration in the brain. Thus, the RAN translation-related proteins containing polyS and polyL have the potential to propagate and the proteins generated by all polyQ diseases might exert universal toxicity in the recipient cells.


Subject(s)
Astrocytes/drug effects , Brain/drug effects , Neurogenesis/drug effects , Neurons/drug effects , Peptides/toxicity , Animals , Astrocytes/metabolism , Astrocytes/ultrastructure , Behavior, Animal/drug effects , Brain/metabolism , Brain/ultrastructure , Cell Death/drug effects , Elevated Plus Maze Test , Locomotion/drug effects , Mice, Inbred ICR , Neurons/metabolism , Neurons/ultrastructure , PC12 Cells , Peptides/metabolism , Rats , Swimming
4.
Ultrasonics ; 118: 106580, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34555738

ABSTRACT

Recently, a method for estimating three-dimensional acoustic impedance profiles in cultured cells and human dermal organs was proposed by interpreting the reflected ultrasonic signal based on a 1-D transmission line model for acoustic impedance microscopy (AIM). However, AIM has a disadvantage that reflected signals from cells overlap with that from a reference substrate. Additionally, the amplitudes of the reflected signals from the specimens are significantly weaker than that from the substrate. In this paper, we proposed a new method for separation of those signals based on a concept of clutter filter, which had been developed for a color Doppler method in medical ultrasonic imaging. The proposed filter using singular value decomposition (SVD) could separate original signals into desired signals such as those from the substrate and cells. Additionally, an effect from a tilt of the substrate was investigated in this study. Separability of the proposed filter was evaluated by two investigations. First one was to evaluate the separability by estimating a correlation coefficient between the filtered signal and signal reflected from a position only with the substrate. Second one was to compare a slope of the substrate estimated from the original signal with that estimated from the filtered signals from the substrate. The experimental results showed that the proposed filter could separate signals from the substrate, and the compensation of the tilt of the substrate could improve the performance of the proposed filter.


Subject(s)
Astrocytes/ultrastructure , Cells, Cultured/ultrastructure , Microscopy, Acoustic/instrumentation , Animals , Equipment Design , Rats , Signal Processing, Computer-Assisted
5.
Alcohol Clin Exp Res ; 46(1): 29-39, 2022 01.
Article in English | MEDLINE | ID: mdl-34839533

ABSTRACT

BACKGROUND: Chloride intracellular channel 4 (CLIC4) is a multifunctional metamorphic protein for which a growing body of evidence supports a major role in the brain's molecular and behavioral responses to ethanol (EtOH). Although key to understanding the functional biology underlying this role, little is known about the cellular and subcellular expression patterns of CLIC4 in brain and how they are affected by EtOH. METHODS: We used qRT-PCR to assess Clic4 mRNA expression in the medial prefrontal cortex (mPFC) of C57BL/6J mice in the absence and presence of acute EtOH exposure. Two complementary immunohistochemical techniques were employed to assess the subcellular localization of the CLIC4 protein and its pattern of expression across brain cell types in the mPFC in the absence and presence of acute EtOH. RESULTS: Through immunohistochemical and stereological techniques, we show that CLIC4 protein is robustly expressed by oligodendrocytes (most abundant), microglia, and astrocytes, with minimal expression in neurons. Following acute EtOH exposure, we observed a rapid increase in Clic4 mRNA expression in female but not male mice and an overall increase in the number of oligodendrocytes and astrocytes expressing the CLIC4 protein. CONCLUSIONS: These findings suggest that Clic4 functions as an early response gene for acute EtOH in brain, which likely underlies its ability to modulate EtOH behavior. Our results also suggest that the role of CLIC4 in the brain's response to EtOH is mediated through oligodendrocytes.


Subject(s)
Chloride Channels/genetics , Ethanol/pharmacology , Gene Expression Regulation/drug effects , Mitochondrial Proteins/genetics , Prefrontal Cortex/metabolism , Transcriptome/drug effects , Animals , Astrocytes/metabolism , Astrocytes/ultrastructure , Behavior, Animal/drug effects , Chloride Channels/analysis , Chloride Channels/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/analysis , Mitochondrial Proteins/physiology , Oligodendroglia/metabolism , Prefrontal Cortex/chemistry , Prefrontal Cortex/drug effects , RNA, Messenger/analysis , Sex Characteristics
6.
Biomolecules ; 11(10)2021 10 06.
Article in English | MEDLINE | ID: mdl-34680100

ABSTRACT

Astrocytes are complex glial cells that play many essential roles in the brain, including the fine-tuning of synaptic activity and blood flow. These roles are linked to fluctuations in intracellular Ca2+ within astrocytes. Recent advances in imaging techniques have identified localized Ca2+ transients within the fine processes of the astrocytic structure, which we term microdomain Ca2+ events. These Ca2+ transients are very diverse and occur under different conditions, including in the presence or absence of surrounding circuit activity. This complexity suggests that different signalling mechanisms mediate microdomain events which may then encode specific astrocyte functions from the modulation of synapses up to brain circuits and behaviour. Several recent studies have shown that a subset of astrocyte microdomain Ca2+ events occur rapidly following local neuronal circuit activity. In this review, we consider the physiological relevance of microdomain astrocyte Ca2+ signalling within brain circuits and outline possible pathways of extracellular Ca2+ influx through ionotropic receptors and other Ca2+ ion channels, which may contribute to astrocyte microdomain events with potentially fast dynamics.


Subject(s)
Astrocytes/cytology , Calcium Signaling/genetics , Calcium/metabolism , Synapses/genetics , Astrocytes/physiology , Astrocytes/ultrastructure , Blood Circulation/genetics , Brain/metabolism , Brain/ultrastructure , Humans , Neuroglia/metabolism , Neuroglia/ultrastructure , Synapses/ultrastructure
7.
Dis Model Mech ; 14(10)2021 10 01.
Article in English | MEDLINE | ID: mdl-34524402

ABSTRACT

Amyloid ß (Aß) peptides generated from the amyloid precursor protein (APP) play a critical role in the development of Alzheimer's disease (AD) pathology. Aß-containing neuronal exosomes, which represent a novel form of intercellular communication, have been shown to influence the function/vulnerability of neurons in AD. Unlike neurons, the significance of exosomes derived from astrocytes remains unclear. In this study, we evaluated the significance of exosomes derived from U18666A-induced cholesterol-accumulated astrocytes in the development of AD pathology. Our results show that cholesterol accumulation decreases exosome secretion, whereas lowering cholesterol increases exosome secretion, from cultured astrocytes. Interestingly, exosomes secreted from U18666A-treated astrocytes contain higher levels of APP, APP-C-terminal fragments, soluble APP, APP secretases and Aß1-40 than exosomes secreted from control astrocytes. Furthermore, we show that exosomes derived from U18666A-treated astrocytes can lead to neurodegeneration, which is attenuated by decreasing Aß production or by neutralizing exosomal Aß peptide with an anti-Aß antibody. These results, taken together, suggest that exosomes derived from cholesterol-accumulated astrocytes can play an important role in trafficking APP/Aß peptides and influencing neuronal viability in the affected regions of the AD brain.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Astrocytes/metabolism , Cholesterol/metabolism , Exosomes/metabolism , Amyloid beta-Peptides/metabolism , Androstenes/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/ultrastructure , Autophagy/drug effects , Cathepsin D/metabolism , Cell Survival/drug effects , Cells, Cultured , Exosomes/drug effects , Exosomes/ultrastructure , Female , Lysosomal-Associated Membrane Protein 1 , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/ultrastructure , Mice, Inbred BALB C , Microtubule-Associated Proteins , Neurons/drug effects , Neurons/metabolism , Rats
8.
Elife ; 102021 09 30.
Article in English | MEDLINE | ID: mdl-34590578

ABSTRACT

Astrocytes are essential cells of the central nervous system, characterized by dynamic relationships with neurons that range from functional metabolic interactions and regulation of neuronal firing activities, to the release of neurotrophic and neuroprotective factors. In Parkinson's disease (PD), dopaminergic neurons are progressively lost during the course of the disease, but the effects of PD on astrocytes and astrocyte-to-neuron communication remain largely unknown. This study focuses on the effects of the PD-related mutation LRRK2 G2019S in astrocytes generated from patient-derived induced pluripotent stem cells. We report the alteration of extracellular vesicle (EV) biogenesis in astrocytes and identify the abnormal accumulation of key PD-related proteins within multivesicular bodies (MVBs). We found that dopaminergic neurons internalize astrocyte-secreted EVs and that LRRK2 G2019S EVs are abnormally enriched in neurites and fail to provide full neurotrophic support to dopaminergic neurons. Thus, dysfunctional astrocyte-to-neuron communication via altered EV biological properties may participate in the progression of PD.


Subject(s)
Astrocytes/enzymology , Cell Communication , Dopaminergic Neurons/enzymology , Exosomes/enzymology , Induced Pluripotent Stem Cells/enzymology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Neural Stem Cells/enzymology , Parkinson Disease/enzymology , Animals , Astrocytes/ultrastructure , Atrophy , Case-Control Studies , Cell Line , Dopaminergic Neurons/pathology , Endocytosis , Exosomes/genetics , Exosomes/ultrastructure , Humans , Induced Pluripotent Stem Cells/ultrastructure , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Neural Stem Cells/ultrastructure , Organelle Biogenesis , Parkinson Disease/genetics , Parkinson Disease/pathology
9.
J Neurosci ; 41(34): 7171-7181, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34253626

ABSTRACT

Mediator protein complex subunit 12 (Med12) is a core component of the basal transcriptional apparatus and plays a critical role in the development of many tissues. Mutations in Med12 are associated with X-linked intellectual disability syndromes and hearing loss; however, its role in nervous system function remains undefined. Here, we show that temporal conditional deletion of Med12 in astrocytes in the adult CNS results in region-specific alterations in astrocyte morphology. Surprisingly, behavioral studies revealed rapid hearing loss after adult deletion of Med12 that was confirmed by a complete abrogation of auditory brainstem responses. Cellular analysis of the cochlea revealed degeneration of the stria vascularis, in conjunction with disorganization of basal cells adjacent to the spiral ligament and downregulation of key cell adhesion proteins. Physiologic analysis revealed early changes in endocochlear potential, consistent with strial-specific defects. Together, our studies reveal that Med12 regulates auditory function in the adult by preserving the structural integrity of the stria vascularis.SIGNIFICANCE STATEMENT Mutations in Mediator protein complex subunit 12 (Med12) are associated with X-linked intellectual disability syndromes and hearing loss. Using temporal-conditional genetic approaches in CNS glia, we found that loss of Med12 results in severe hearing loss in adult animals through rapid degeneration of the stria vascularis. Our study describes the first animal model that recapitulates hearing loss identified in Med12-related disorders and provides a new system in which to examine the underlying cellular and molecular mechanisms of Med12 function in the adult nervous system.


Subject(s)
Astrocytes/physiology , Hearing Loss, Sensorineural/etiology , Mediator Complex/deficiency , Stria Vascularis/pathology , Animals , Astrocytes/metabolism , Astrocytes/ultrastructure , Cell Adhesion Molecules/metabolism , Conditioning, Classical/physiology , Evoked Potentials, Auditory, Brain Stem , Fear , Female , Freezing Reaction, Cataleptic , Gene Knockout Techniques , Hearing Loss, Sensorineural/pathology , Hearing Loss, Sensorineural/physiopathology , Male , Mediator Complex/physiology , Mice , Organ Specificity , Otoacoustic Emissions, Spontaneous , Random Allocation , Reflex, Startle
10.
Aging (Albany NY) ; 13(12): 15917-15941, 2021 06 19.
Article in English | MEDLINE | ID: mdl-34139671

ABSTRACT

The G-quadruplex (G4-DNA or G4) is a secondary DNA structure formed by DNA sequences containing multiple runs of guanines. While it is now firmly established that stabilized G4s lead to enhanced genomic instability in cancer cells, whether and how G4s contribute to genomic instability in brain cells is still not clear. We previously showed that, in cultured primary neurons, small-molecule G4 stabilizers promote formation of DNA double-strand breaks (DSBs) and downregulate the Brca1 gene. Here, we determined if G4-dependent Brca1 downregulation is unique to neurons or if the effects in neurons also occur in astrocytes and microglia. We show that primary neurons, astrocytes and microglia basally exhibit different G4 landscapes. Stabilizing G4-DNA with the G4 ligand pyridostatin (PDS) differentially modifies chromatin structure in these cell types. Intriguingly, PDS promotes DNA DSBs in neurons, astrocytes and microglial cells, but fails to downregulate Brca1 in astrocytes and microglia, indicating differences in DNA damage and repair pathways between brain cell types. Taken together, our findings suggest that stabilized G4-DNA contribute to genomic instability in the brain and may represent a novel senescence pathway in brain aging.


Subject(s)
Astrocytes/metabolism , G-Quadruplexes , Microglia/metabolism , Neurons/metabolism , Aminoquinolines/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/ultrastructure , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Base Sequence , Cell Line , Chromatin/drug effects , Chromatin/metabolism , Chromatin/ultrastructure , DNA Damage , Mice , Microglia/drug effects , Microglia/ultrastructure , Neurons/drug effects , Neurons/ultrastructure , Picolinic Acids/pharmacology , Promoter Regions, Genetic/genetics , Rats
11.
Cell Death Dis ; 12(3): 225, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33649354

ABSTRACT

Conversion of astrocytes into neurons in vivo offers an alternative therapeutic approach for neuronal loss after injury or disease. However, not only the efficiency of the conversion of astrocytes into functional neurons by single Neurog2, but also the conundrum that whether Neurog2-induced neuronal cells (Neurog2-iNs) are further functionally integrated into existing matured neural circuits remains unknown. Here, we adopted the AAV(2/8) delivery system to overexpress single factor Neurog2 into astrocytes and found that the majority of astrocytes were successfully converted into neuronal cells in multiple brain regions, including the midbrain and spinal cord. In the midbrain, Neurog2-induced neuronal cells (Neurog2-iNs) exhibit neuronal morphology, mature electrophysiological properties, glutamatergic identity (about 60%), and synapse-like configuration local circuits. In the spinal cord, astrocytes from both the intact and lesioned sources could be converted into functional neurons with ectopic expression of Neurog2 alone. Notably, further evidence from our study also proves that Neurog2-iNs in the intact spinal cord are capable of responding to diverse afferent inputs from dorsal root ganglion (DRG). Together, this study does not merely demonstrate the feasibility of Neurog2 for efficient in vivo reprogramming, it gives an indication for the Neurog2-iNs as a functional and potential factor in cell-replacement therapy.


Subject(s)
Astrocytes/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Transdifferentiation , Mesencephalon/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis , Neurons/metabolism , Spinal Cord/metabolism , Animals , Astrocytes/ultrastructure , Basic Helix-Loop-Helix Transcription Factors/genetics , Cells, Cultured , Dependovirus/genetics , Gene Transfer Techniques , Genetic Vectors , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Mesencephalon/ultrastructure , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/ultrastructure , Oxidoreductases Acting on CH-NH Group Donors/genetics , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Phenotype , Spinal Cord/ultrastructure , Vesicular Glutamate Transport Protein 2/genetics , Vesicular Glutamate Transport Protein 2/metabolism
12.
J Neurochem ; 156(6): 848-866, 2021 03.
Article in English | MEDLINE | ID: mdl-32939791

ABSTRACT

This study aimed to investigate and compare cell growth manners and functional differences of primary cortical neurons cultured on either poly-d-lysine (PDL) and or Matrigel, to delineate the role of extracellular matrix on providing resemblance to in vivo cellular interactions in nervous tissue. Primary cortical neurons, obtained from embryonic day 15 mice pups, seeded either on PDL- or Matrigel-coated culture ware were investigated by DIC/bright field and fluorescence/confocal microscopy for their morphology, 2D and 3D structure, and distribution patterns. Patch clamp, western blot, and RT-PCR studies were performed to investigate neuronal firing thresholds and sodium channel subtypes Nav1.2 and Nav1.6 expression. Cortical neurons cultured on PDL coating possessed a 2D structure composed of a few numbers of branched and tortuous neurites that contacted with each other in one to one manner, however, neurons on Matrigel coating showed a more complicated dimensional network that depicted tight, linear axonal bundles forming a 3D interacted neuron-astrocyte construction. This difference in growth patterns also showed a significant alteration in neuronal firing threshold which was recorded between 80 < Iinj > 120 pA on PDL and 2 < Iinj > 160 pA on Matrigel. Neurons grown up on Matrigel showed increased levels of sodium channel protein expression of Nav1.2 and Nav1.6 compared to neurons on PDL. These results have demonstrated that a 3D interacted neuron-astrocyte construction on Matrigel enhances the development of Nav1.2 and Nav1.6 in vitro and decreases neuronal firing threshold by 40 times compared to conventional PDL, resembling in vivo neuronal networks and hence would be a better in vitro model of adult neurons.


Subject(s)
Astrocytes/physiology , Astrocytes/ultrastructure , Collagen , Laminin , Neurons/physiology , Neurons/ultrastructure , Proteoglycans , Voltage-Gated Sodium Channels/biosynthesis , Animals , Cerebral Cortex/cytology , Drug Combinations , Electrophysiological Phenomena , Embryo, Mammalian/physiology , Female , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , NAV1.2 Voltage-Gated Sodium Channel/biosynthesis , NAV1.2 Voltage-Gated Sodium Channel/genetics , NAV1.6 Voltage-Gated Sodium Channel/biosynthesis , NAV1.6 Voltage-Gated Sodium Channel/genetics , Neurites/physiology , Patch-Clamp Techniques , Pregnancy , Primary Cell Culture
13.
J Neurosci Res ; 99(2): 467-480, 2021 02.
Article in English | MEDLINE | ID: mdl-33105056

ABSTRACT

Idiopathic intracranial hypertension (IIH) primarily affects fertile, overweight women, and presents with the symptoms of raised intracranial pressure. The etiology is unknown but has been thought to relate to cerebrospinal fluid disturbance or cerebral venous stenosis. We have previously found evidence that IIH is also a disease of the brain parenchyma, evidenced by alterations at the neurogliovascular interface, including astrogliosis, pathological changes in the basement membrane and pericytes, and alterations of perivascular aquaporin-4. The aim of this present electron microscopic study was to examine whether mitochondria phenotype was changed in IIH, particularly focusing on perivascular astrocytic endfeet and neurons (soma and pre- and postsynaptic terminals). Cortical brain biopsies of nine reference individuals and eight IIH patients were analyzed for subcellular distribution and phenotypical features of mitochondria using transmission electron microscopy. We found significantly increased prevalence of pathological mitochondria and reduced number of normal mitochondria in astrocytic endfeet of IIH patients. The degree of astrogliosis correlated negatively with the number of normal mitochondria in astrocytic endfoot processes. Moreover, we found significantly increased number of pathological mitochondria in pre- and postsynaptic neuronal terminals, as well as significantly shortened distance between mitochondria and endoplasmic reticulum contacts. Finally, the length of postsynaptic density, a marker of synaptic strength, was on average reduced in IIH. The present data provide evidence of pathological mitochondria in perivascular astrocytes endfeet and neurons of IIH patients, highlighting that impaired metabolism at the neurogliovascular interface may be a facet of IIH.


Subject(s)
Astrocytes/ultrastructure , Cerebral Cortex/pathology , Mitochondria/pathology , Neurons/ultrastructure , Pseudotumor Cerebri/pathology , Adult , Biopsy , Endoplasmic Reticulum/ultrastructure , Female , Gliosis/etiology , Gliosis/pathology , Glymphatic System/ultrastructure , Humans , Male , Microscopy, Electron , Middle Aged , Nerve Endings/ultrastructure , Post-Synaptic Density/ultrastructure , Prospective Studies , Pseudotumor Cerebri/complications , Single-Blind Method , Young Adult
14.
Int J Mol Sci ; 21(23)2020 Nov 26.
Article in English | MEDLINE | ID: mdl-33256007

ABSTRACT

Due to strong antimicrobial properties, silver nanoparticles (AgNPs) are used in a wide range of medical and consumer products, including those dedicated for infants and children. While AgNPs are known to exert neurotoxic effects, current knowledge concerning their impact on the developing brain is scarce. During investigations of mechanisms of neurotoxicity in immature rats, we studied the influence of AgNPs on glutamate transporter systems which are involved in regulation of extracellular concentration of glutamate, an excitotoxic amino acid, and compared it with positive control-Ag citrate. We identified significant deposition of AgNPs in brain tissue of exposed rats over the post-exposure time. Ultrastructural alterations in endoplasmic reticulum (ER) and Golgi complexes were observed in neurons of AgNP-exposed rats, which are characteristics of ER stress. These changes presumably underlie substantial long-lasting downregulation of neuronal glutamate transporter EAAC1, which was noted in AgNP-exposed rats. Conversely, the expression of astroglial glutamate transporters GLT-1 and GLAST was not affected by exposure to AgNPs, but the activity of the transporters was diminished. These results indicate that even low doses of AgNPs administered during an early stage of life create a substantial risk for health of immature organisms. Hence, the safety of AgNP-containing products for infants and children should be carefully considered.


Subject(s)
Amino Acid Transport System X-AG/metabolism , Brain/metabolism , Metal Nanoparticles/toxicity , Silver/toxicity , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/ultrastructure , Brain/drug effects , Excitatory Amino Acid Transporter 3/metabolism , Glutamic Acid/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/ultrastructure , Rats , Silver/blood , Sodium/metabolism , Time Factors
15.
Cells ; 9(11)2020 11 05.
Article in English | MEDLINE | ID: mdl-33167342

ABSTRACT

Aquaporin-4 (AQP4) is critically involved in brain water and volume homeostasis and has been implicated in a wide range of pathological conditions. Notably, evidence has been accrued to suggest that AQP4 plays a proinflammatory role by promoting release of astrocytic cytokines that activate microglia and other astrocytes. Neuroinflammation is a hallmark of Parkinson's disease (PD), and we have previously shown that astrocytes in substantia nigra (SN) are enriched in AQP4 relative to cortical astrocytes, and that their complement of AQP4 is further increased following treatment with the parkinsonogenic toxin MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine). Here, we investigated the effect of Aqp4 deletion on microglial activation in mice subjected to unilateral intrastriatal injection of 1-methyl-4-phenylpyridinium (MPP+, the toxic metabolite of MPTP). Our results show that MPP+ injections lead to a pronounced increase in the expression level of microglial activating genes in the ventral mesencephalon of wild type (WT) mice, but not Aqp4-/- mice. We also show, in WT mice, that MPP+ injections cause an upregulation of nigral AQP4 and swelling of astrocytic endfeet. These findings are consistent with the idea that AQP4 plays a pro-inflammatory role in Parkinson's disease, secondary to the dysregulation of astrocytic volume homeostasis.


Subject(s)
1-Methyl-4-phenylpyridinium/administration & dosage , Aquaporin 4/metabolism , Inflammation/metabolism , Inflammation/pathology , Parkinson Disease/pathology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Astrocytes/ultrastructure , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Female , Gene Expression Regulation , Glial Fibrillary Acidic Protein/metabolism , Injections , Male , Mesencephalon/pathology , Mice, Inbred C57BL , Neuroglia/pathology , Parkinson Disease/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Substantia Nigra/pathology
16.
Med Sci Monit ; 26: e925754, 2020 Oct 09.
Article in English | MEDLINE | ID: mdl-33077704

ABSTRACT

BACKGROUND With infiltration, high-grade glioma easily causes the boundary between tumor tissue and adjacent tissue to become unclear and results in tumor recurrence at or near the resection margin according to the incomplete surgical resection. Fourier transform infrared spectroscopy (FTIR) technique has been demonstrated to be a useful tool that yields a molecular fingerprint and provides rapid, nondestructive, high-throughput and clinically relevant diagnostic information. MATERIAL AND METHODS FTIR was used to investigate the morphological and biochemical properties of human astrocytes (HA), microglia (HM1900), glioma cells (U87), and glioblastoma cells (BT325) cultured in vitro to simulate the infiltration area, with the use of multi-peak fitting and principal component analysis (PCA) of amide I of FTIR spectra and the use of hierarchical cluster analysis (HCA). RESULTS We found that the secondary structures of the 4 types of cells were significantly different. The contents of a-helix structure in glial cells was significantly higher than in the glioma cells, but the levels of ß-sheet, ß-turn, and random coil structures were lower. The 4 types of cells could be clearly separated with 85% for PC1 and 12.2% for PC2. CONCLUSIONS FTIR can be used to distinguish between human astrocytes, microglia, glioma, and glioblastoma cells in vitro. The protein secondary structure can be used as an indicator to distinguish tumor cells from glial cells. Further tissue-based and in vivo studies are needed to determine whether FTIR can identify cerebral glioma.


Subject(s)
Astrocytes/ultrastructure , Glioblastoma/ultrastructure , Microglia/ultrastructure , Spectroscopy, Fourier Transform Infrared/methods , Astrocytes/cytology , Cell Line, Tumor , Glioblastoma/pathology , Humans , Microglia/cytology
17.
Neuron ; 108(5): 919-936.e11, 2020 12 09.
Article in English | MEDLINE | ID: mdl-32976770

ABSTRACT

Extrasynaptic actions of glutamate are limited by high-affinity transporters expressed by perisynaptic astroglial processes (PAPs): this helps maintain point-to-point transmission in excitatory circuits. Memory formation in the brain is associated with synaptic remodeling, but how this affects PAPs and therefore extrasynaptic glutamate actions is poorly understood. Here, we used advanced imaging methods, in situ and in vivo, to find that a classical synaptic memory mechanism, long-term potentiation (LTP), triggers withdrawal of PAPs from potentiated synapses. Optical glutamate sensors combined with patch-clamp and 3D molecular localization reveal that LTP induction thus prompts spatial retreat of astroglial glutamate transporters, boosting glutamate spillover and NMDA-receptor-mediated inter-synaptic cross-talk. The LTP-triggered PAP withdrawal involves NKCC1 transporters and the actin-controlling protein cofilin but does not depend on major Ca2+-dependent cascades in astrocytes. We have therefore uncovered a mechanism by which a memory trace at one synapse could alter signal handling by multiple neighboring connections.


Subject(s)
Astrocytes/metabolism , Glutamic Acid/metabolism , Long-Term Potentiation/physiology , Synapses/metabolism , Animals , Astrocytes/ultrastructure , Female , Imaging, Three-Dimensional/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Rats, Wistar , Synapses/ultrastructure
18.
Neuron ; 108(4): 691-706.e10, 2020 11 25.
Article in English | MEDLINE | ID: mdl-32905785

ABSTRACT

Sensory discrimination is essential for survival. However, how sensory information is finely controlled in the brain is not well defined. Here, we show that astrocytes control tactile acuity via tonic inhibition in the thalamus. Mechanistically, diamine oxidase (DAO) and the subsequent aldehyde dehydrogenase 1a1 (Aldh1a1) convert putrescine into GABA, which is released via Best1. The GABA from astrocytes inhibits synaptically evoked firing at the lemniscal synapses to fine-tune the dynamic range of the stimulation-response relationship, the precision of spike timing, and tactile discrimination. Our findings reveal a novel role of astrocytes in the control of sensory acuity through tonic GABA release.


Subject(s)
Astrocytes/physiology , Neural Inhibition/physiology , Thalamus/physiology , Touch Perception/physiology , gamma-Aminobutyric Acid/physiology , Aldehyde Dehydrogenase 1 Family/metabolism , Amine Oxidase (Copper-Containing)/metabolism , Animals , Astrocytes/metabolism , Astrocytes/ultrastructure , Bestrophins/biosynthesis , Bestrophins/genetics , Female , GABA Antagonists , Immunohistochemistry , Inhibitory Postsynaptic Potentials/physiology , Macrolides/pharmacology , Male , Mice , Mice, Knockout , Microscopy, Electron , Neurons/metabolism , Neurons/physiology , Patch-Clamp Techniques , Picrotoxin/pharmacology , Primary Cell Culture , Pyridazines/pharmacology , RNA, Small Interfering/pharmacology , Retinal Dehydrogenase/metabolism , gamma-Aminobutyric Acid/biosynthesis , gamma-Aminobutyric Acid/pharmacology
19.
Neuron ; 108(1): 111-127.e6, 2020 10 14.
Article in English | MEDLINE | ID: mdl-32795398

ABSTRACT

Cajal recognized that the elaborate shape of neurons is fundamental to their function in the brain. However, there are no simple and generalizable genetic methods to study neuronal or glial cell morphology in the mammalian brain. Here, we describe four mouse lines conferring Cre-dependent sparse cell labeling based on mononucleotide repeat frameshift (MORF) as a stochastic translational switch. Notably, the optimized MORF3 mice, with a membrane-bound multivalent immunoreporter, confer Cre-dependent sparse and bright labeling of thousands of neurons, astrocytes, or microglia in each brain, revealing their intricate morphologies. MORF3 mice are compatible with imaging in tissue-cleared thick brain sections and with immuno-EM. An analysis of 151 MORF3-labeled developing retinal horizontal cells reveals novel morphological cell clusters and axonal maturation patterns. Our study demonstrates a conceptually novel, simple, generalizable, and scalable mouse genetic solution to sparsely label and illuminate the morphology of genetically defined neurons and glia in the mammalian brain.


Subject(s)
Astrocytes/ultrastructure , Brain/ultrastructure , Microglia/ultrastructure , Neurons/ultrastructure , Retinal Horizontal Cells/ultrastructure , Animals , Astrocytes/metabolism , Astrocytes/pathology , Brain/metabolism , Brain/pathology , Frameshift Mutation/genetics , Green Fluorescent Proteins/genetics , Integrases , Mice , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Microsatellite Repeats/genetics , Neurons/metabolism , Neurons/pathology , Retinal Horizontal Cells/metabolism , Retinal Horizontal Cells/pathology
20.
Cells ; 9(7)2020 07 02.
Article in English | MEDLINE | ID: mdl-32630739

ABSTRACT

Despite the remarkable complexity of the individual neuron and of neuronal circuits, it has been clear for quite a while that, in order to understand the functioning of the brain, the contribution of other cell types in the brain have to be accounted for. Among glial cells, astrocytes have multiple roles in orchestrating neuronal functions. Their communication with neurons by exchanging signaling molecules and removing molecules from extracellular space takes place at several levels and is governed by different cellular processes, supported by multiple cellular structures, including the cytoskeleton. Intermediate filaments in astrocytes are emerging as important integrators of cellular processes. Astrocytes express five types of intermediate filaments: glial fibrillary acidic protein (GFAP); vimentin; nestin; synemin; lamins. Variability, interactions with different cellular structures and the particular roles of individual intermediate filaments in astrocytes have been studied extensively in the case of GFAP and vimentin, but far less attention has been given to nestin, synemin and lamins. Similarly, the interplay between different types of cytoskeleton and the interaction between the cytoskeleton and membranous structures, which is mediated by cytolinker proteins, are understudied in astrocytes. The present review summarizes the basic properties of astrocytic intermediate filaments and of other cytoskeletal macromolecules, such as cytolinker proteins, and describes the current knowledge of their roles in normal physiological and pathological conditions.


Subject(s)
Astrocytes/metabolism , Intermediate Filaments/metabolism , Animals , Astrocytes/ultrastructure , Humans , Intermediate Filament Proteins/chemistry , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Intermediate Filaments/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...