Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Nat Metab ; 6(3): 494-513, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38443593

ABSTRACT

Long-lasting pain stimuli can trigger maladaptive changes in the spinal cord, reminiscent of plasticity associated with memory formation. Metabolic coupling between astrocytes and neurons has been implicated in neuronal plasticity and memory formation in the central nervous system, but neither its involvement in pathological pain nor in spinal plasticity has been tested. Here we report a form of neuroglia signalling involving spinal astrocytic glycogen dynamics triggered by persistent noxious stimulation via upregulation of the Protein Targeting to Glycogen (PTG) in spinal astrocytes. PTG drove glycogen build-up in astrocytes, and blunting glycogen accumulation and turnover by Ptg gene deletion reduced pain-related behaviours and promoted faster recovery by shortening pain maintenance in mice. Furthermore, mechanistic analyses revealed that glycogen dynamics is a critically required process for maintenance of pain by facilitating neuronal plasticity in spinal lamina 1 neurons. In summary, our study describes a previously unappreciated mechanism of astrocyte-neuron metabolic communication through glycogen breakdown in the spinal cord that fuels spinal neuron hyperexcitability.


Subject(s)
Astrocytes , Pain , Mice , Animals , Astrocytes/metabolism , Pain/metabolism , Pain/pathology , Neurons/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Glycogen/metabolism
2.
J Neuroinflammation ; 19(1): 279, 2022 Nov 19.
Article in English | MEDLINE | ID: mdl-36403069

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) is characterized by massive changes in neuronal excitation, from acute excitotoxicity to chronic hyper- or hypoexcitability. Nuclear calcium signaling pathways are involved in translating changes in synaptic inputs and neuronal activity into discrete transcriptional programs which not only affect neuronal survival and synaptic integrity, but also the crosstalk between neurons and glial cells. Here, we report the effects of blunting neuronal nuclear calcium signals in the context of TBI. METHODS: We used AAV vectors to express the genetically encoded and nuclear-targeted calcium buffer parvalbumin (PV.NLS.mCherry) or the calcium/calmodulin buffer CaMBP4.mCherry in neurons only. Upon TBI, the extent of neuroinflammation, neuronal death and synaptic loss were assessed by immunohistochemistry and targeted transcriptome analysis. Modulation of the overall level of neuronal activity was achieved by PSAM/PSEM chemogenetics targeted to parvalbumin interneurons. The functional impact of neuronal nuclear calcium buffering in TBI was assessed by quantification of spontaneous whisking. RESULTS: Buffering neuronal nuclear calcium unexpectedly resulted in a massive and long-lasting increase in the recruitment of reactive microglia to the injury site, which was characterized by a disease-associated and phagocytic phenotype. This effect was accompanied by a substantial surge in synaptic loss and significantly reduced whisking activity. Transcriptome analysis revealed a complex effect of TBI in the context of neuronal nuclear calcium buffering, with upregulation of complement factors, chemokines and interferon-response genes, as well as the downregulation of synaptic genes and epigenetic regulators compared to control conditions. Notably, nuclear calcium buffering led to a substantial loss in neuronal osteoprotegerin (OPG), whereas stimulation of neuronal firing induced OPG expression. Viral re-expression of OPG resulted in decreased microglial recruitment and synaptic loss. OPG upregulation was also observed in the CSF of human TBI patients, underscoring its translational value. CONCLUSION: Neuronal nuclear calcium signals regulate the degree of microglial recruitment and reactivity upon TBI via, among others, osteoprotegerin signals. Our findings support a model whereby neuronal activity altered after TBI exerts a powerful impact on the neuroinflammatory cascade, which in turn contributes to the overall loss of synapses and functional impairment.


Subject(s)
Brain Injuries, Traumatic , Microglia , Humans , Microglia/metabolism , Calcium Signaling , Parvalbumins/metabolism , Calcium/metabolism , Osteoprotegerin/metabolism , Brain Injuries, Traumatic/metabolism
3.
Nat Cell Biol ; 24(9): 1407-1421, 2022 09.
Article in English | MEDLINE | ID: mdl-36097071

ABSTRACT

Mechanistic target of rapamycin complex 1 (mTORC1) senses nutrient availability to appropriately regulate cellular anabolism and catabolism. During nutrient restriction, different organs in an animal do not respond equally, with vital organs being relatively spared. This raises the possibility that mTORC1 is differentially regulated in different cell types, yet little is known about this mechanistically. The Rag GTPases, RagA or RagB bound to RagC or RagD, tether mTORC1 in a nutrient-dependent manner to lysosomes where mTORC1 becomes activated. Although the RagA and B paralogues were assumed to be functionally equivalent, we find here that the RagB isoforms, which are highly expressed in neurons, impart mTORC1 with resistance to nutrient starvation by inhibiting the RagA/B GTPase-activating protein GATOR1. We further show that high expression of RagB isoforms is observed in some tumours, revealing an alternative strategy by which cancer cells can retain elevated mTORC1 upon low nutrient availability.


Subject(s)
Multiprotein Complexes , Signal Transduction , Animals , Brain/metabolism , GTPase-Activating Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism
4.
EMBO Rep ; 23(9): e54078, 2022 09 05.
Article in English | MEDLINE | ID: mdl-35861333

ABSTRACT

According to the current consensus, murine neural stem cells (NSCs) apically contacting the lateral ventricle generate differentiated progenitors by rare asymmetric divisions or by relocating to the basal side of the ventricular-subventricular zone (V-SVZ). Both processes will ultimately lead to the generation of adult-born olfactory bulb (OB) interneurons. In contrast to this view, we here find that adult-born OB interneurons largely derive from an additional NSC-type resident in the basal V-SVZ. Despite being both capable of self-renewal and long-term quiescence, apical and basal NSCs differ in Nestin expression, primary cilia extension and frequency of cell division. The expression of Notch-related genes also differs between the two NSC groups, and Notch activation is greatest in apical NSCs. Apical downregulation of Notch-effector Hes1 decreases Notch activation while increasing proliferation across the niche and neurogenesis from apical NSCs. Underscoring their different roles in neurogenesis, lactation-dependent increase in neurogenesis is paralleled by extra activation of basal but not apical NSCs. Thus, basal NSCs support OB neurogenesis, whereas apical NSCs impart Notch-mediated lateral inhibition across the V-SVZ.


Subject(s)
Lateral Ventricles , Neural Stem Cells , Animals , Cell Differentiation/genetics , Female , Lateral Ventricles/metabolism , Mice , Neural Stem Cells/metabolism , Neurogenesis/genetics , Olfactory Bulb/metabolism
5.
Nat Commun ; 13(1): 875, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35169129

ABSTRACT

Persistent pain is sustained by maladaptive changes in gene transcription resulting in altered function of the relevant circuits; therapies are still unsatisfactory. The epigenetic mechanisms and affected genes linking nociceptive activity to transcriptional changes and pathological sensitivity are unclear. Here, we found that, among several histone deacetylases (HDACs), synaptic activity specifically affects HDAC4 in murine spinal cord dorsal horn neurons. Noxious stimuli that induce long-lasting inflammatory hypersensitivity cause nuclear export and inactivation of HDAC4. The development of inflammation-associated mechanical hypersensitivity, but neither acute nor basal sensitivity, is impaired by the expression of a constitutively nuclear localized HDAC4 mutant. Next generation RNA-sequencing revealed an HDAC4-regulated gene program comprising mediators of sensitization including the organic anion transporter OAT1, known for its renal transport function. Using pharmacological and molecular tools to modulate OAT1 activity or expression, we causally link OAT1 to persistent inflammatory hypersensitivity in mice. Thus, HDAC4 is a key epigenetic regulator that translates nociceptive activity into sensitization by regulating OAT1, which is a potential target for pain-relieving therapies.


Subject(s)
Chronic Pain/pathology , Histone Deacetylases/metabolism , Neuralgia/pathology , Nociceptive Pain/pathology , Organic Anion Transport Protein 1/metabolism , Spinal Cord Dorsal Horn/metabolism , Animals , Cells, Cultured , Dependovirus/genetics , Female , Hypersensitivity/pathology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Organic Anion Transport Protein 1/antagonists & inhibitors , RNA Interference , RNA, Small Interfering/genetics , Spinal Cord Dorsal Horn/cytology
6.
J Biol Chem ; 298(2): 101508, 2022 02.
Article in English | MEDLINE | ID: mdl-34942149

ABSTRACT

The mitochondrial solute carrier family 8 sodium/calcium/lithium exchanger, member B1 (NCLX) is an important mediator of calcium extrusion from mitochondria. In this study, we tested the hypothesis that physiological expression levels of NCLX are essential for maintaining neuronal resilience in the face of excitotoxic challenge. Using an shRNA-mediated approach, we showed that reduced NCLX expression exacerbates neuronal mitochondrial calcium dysregulation, mitochondrial membrane potential (ΔΨm) breakdown, and reactive oxygen species generation during excitotoxic stimulation of primary hippocampal cultures. Moreover, NCLX knockdown-which affected both neurons and glia-resulted not only in enhanced neurodegeneration following an excitotoxic insult but also in neuronal and astrocytic cell death under basal conditions. Our data also revealed that synaptic activity, which promotes neuroprotective signaling, can become lethal upon NCLX depletion; expression of NCLX-targeted shRNA impaired the clearance of mitochondrial calcium following action potential bursts, and was associated both with ΔΨm breakdown and substantial neurodegeneration in hippocampal cultures undergoing synaptic activity. Finally, we showed that NCLX knockdown within the hippocampal cornu ammonis 1 region in vivo causes substantial neurodegeneration and astrodegeneration. In summary, we demonstrated that dysregulated NCLX expression not only sensitizes neuroglial networks to excitotoxic stimuli but also notably renders otherwise neuroprotective synaptic activity toxic. These findings may explain the emergence of neurodegeneration and astrodegeneration in patients with disorders characterized by disrupted NCLX expression or function, and suggest that treatments aimed at enhancing or restoring NCLX function may prevent central nervous system damage in these disease states.


Subject(s)
Calcium , Mitochondrial Proteins , Nerve Net , Neuroglia , Sodium-Calcium Exchanger , Calcium/metabolism , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Nerve Net/metabolism , Neuroglia/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sodium-Calcium Exchanger/biosynthesis , Sodium-Calcium Exchanger/genetics , Sodium-Calcium Exchanger/metabolism
7.
Science ; 370(6513)2020 10 09.
Article in English | MEDLINE | ID: mdl-33033186

ABSTRACT

Excitotoxicity induced by NMDA receptors (NMDARs) is thought to be intimately linked to high intracellular calcium load. Unexpectedly, NMDAR-mediated toxicity can be eliminated without affecting NMDAR-induced calcium signals. Instead, excitotoxicity requires physical coupling of NMDARs to TRPM4. This interaction is mediated by intracellular domains located in the near-membrane portions of the receptors. Structure-based computational drug screening using the interaction interface of TRPM4 in complex with NMDARs identified small molecules that spare NMDAR-induced calcium signaling but disrupt the NMDAR/TRPM4 complex. These interaction interface inhibitors strongly reduce NMDA-triggered toxicity and mitochondrial dysfunction, abolish cyclic adenosine monophosphate-responsive element-binding protein (CREB) shutoff, boost gene induction, and reduce neuronal loss in mouse models of stroke and retinal degeneration. Recombinant or small-molecule NMDAR/TRPM4 interface inhibitors may mitigate currently untreatable human neurodegenerative diseases.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Drug Discovery , Neurodegenerative Diseases/metabolism , Neuroprotective Agents/chemistry , Receptors, N-Methyl-D-Aspartate/metabolism , Recombinant Proteins/metabolism , TRPM Cation Channels/metabolism , Animals , Calcium Signaling , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Gene Knockdown Techniques , Humans , Mice , Neurodegenerative Diseases/drug therapy , Protein Domains , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/therapeutic use , Recombinant Proteins/chemistry , Recombinant Proteins/therapeutic use , TRPM Cation Channels/genetics , Transcriptional Activation
8.
Mol Brain ; 13(1): 124, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32928261

ABSTRACT

Glutamate toxicity is a pathomechanism that contributes to neuronal cell death in a wide range of acute and chronic neurodegenerative and neuroinflammatory diseases. Activation of the N-methyl-D-aspartate (NMDA)-type glutamate receptor and breakdown of the mitochondrial membrane potential are key events during glutamate toxicity. Due to its manifold functions in nervous system physiology, however, the NMDA receptor is not well suited as a drug target. To identify novel compounds that act downstream of toxic NMDA receptor signaling and can protect mitochondria from glutamate toxicity, we developed a cell viability screening assay in primary mouse cortical neurons. In a proof-of-principle screen we tested 146 natural products and 424 FDA-approved drugs for their ability to protect neurons against NMDA-induced cell death. We confirmed several known neuroprotective drugs that include Dutasteride, Enalapril, Finasteride, Haloperidol, and Oxybutynin, and we identified neuroprotective properties of Elvitegravir. Using live imaging of tetramethylrhodamine ethyl ester-labelled primary cortical neurons, we found that Elvitegravir, Dutasteride, and Oxybutynin attenuated the NMDA-induced breakdown of the mitochondrial membrane potential. Patch clamp electrophysiological recordings in NMDA receptor-expressing HEK293 cell lines and primary mouse hippocampal neurons revealed that Elvitegravir does not act at the NMDA receptor and does not affect the function of glutamatergic synapses. In summary, we have developed a cost-effective and easy-to-implement screening assay in primary neurons and identified Elvitegravir as a neuro- and mitoprotective drug that acts downstream of the NMDA receptor.


Subject(s)
Antiviral Agents/pharmacology , Drug Approval , Microscopy , Neurons/metabolism , Neuroprotective Agents/pharmacology , Quinolones/pharmacology , Small Molecule Libraries/pharmacology , United States Food and Drug Administration , Animals , Cell Death/drug effects , Cells, Cultured , Channelrhodopsins/metabolism , Excitatory Postsynaptic Potentials/drug effects , HEK293 Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred C57BL , N-Methylaspartate/pharmacology , Neurons/cytology , Neurons/drug effects , Neuroprotection/drug effects , Optogenetics , Receptors, AMPA/metabolism , Receptors, Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , United States
9.
Article in English | MEDLINE | ID: mdl-31570333

ABSTRACT

In the nervous system, calcium signals play a major role in the conversion of synaptic stimuli into transcriptional responses. Signal-regulated gene transcription is fundamental for a range of long-lasting adaptive brain functions that include learning and memory, structural plasticity of neurites and synapses, acquired neuroprotection, chronic pain, and addiction. In this review, we summarize the diverse mechanisms governing calcium-dependent transcriptional regulation associated with central nervous system plasticity. We focus on recent advances in the field of synapse-to-nucleus communication that include studies of the signal-regulated transcriptome in human neurons, identification of novel regulatory mechanisms such as activity-induced DNA double-strand breaks, and the identification of novel forms of activity- and transcription-dependent adaptations, in particular, metabolic plasticity. We summarize the reciprocal interactions between different kinds of neuroadaptations and highlight the emerging role of activity-regulated epigenetic modifiers in gating the inducibility of signal-regulated genes.


Subject(s)
Brain/physiology , Calcium/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , Synapses/metabolism , Transcription, Genetic , Animals , Astrocytes/metabolism , Calcium Signaling , Cell Communication , Cell Line , DNA Breaks, Double-Stranded , Energy Metabolism , Epigenesis, Genetic , Gene Expression Regulation , Glucose/metabolism , Glycolysis , Humans , Memory/physiology , Memory, Long-Term , Mice , Neuronal Plasticity/physiology , Neurons/metabolism , Neuroprotection , Reactive Oxygen Species , Signal Transduction , Substance-Related Disorders
10.
Acta Neuropathol Commun ; 7(1): 15, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30722785

ABSTRACT

Local cerebral hypoperfusion causes ischemic stroke while driving multiple cell-specific responses including inflammation, glutamate-induced neurotoxicity mediated via NMDAR, edema formation and angiogenesis. Despite the relevance of these pathophysiological mechanisms for disease progression and outcome, molecular determinants controlling the onset of these processes are only partially understood. In this context, our study intended to investigate the functional role of EphB2, a receptor tyrosine kinase that is crucial for synapse function and binds to membrane-associated ephrin-B ligands.Cerebral ischemia was induced in Ephb2-/- mice by transient middle cerebral artery occlusion followed by different times (6, 12, 24 and 48 h) of reperfusion. Histological, neurofunctional and transcriptome analyses indicated an increase in EphB2 phosphorylation under these conditions and attenuated progression of stroke in Ephb2-/- mice. Moreover, while infiltration of microglia/macrophages and astrocytes into the peri-infarct region was not altered, expression of the pro-inflammatory mediators MCP-1 and IL-6 was decreased in these mice. In vitro analyses indicated that binding of EphB2 to astrocytic ephrin-B ligands stimulates NF-κB-mediated cytokine expression via the MAPK pathway. Further magnetic resonance imaging of the Ephb2-/- ischemic brain revealed a lower level of cytotoxic edema formation within 6 h upon onset of reperfusion. On the mechanistic level, absence of neuronal EphB2 decreased the mitochondrial Ca2+ load upon specific activation of NMDAR but not during synaptic activity. Furthermore, neuron-specific loss of ephrin-B2 reduced the extent of cerebral tissue damage in the acute phase of ischemic stroke.Collectively, EphB2 may promote the immediate response to an ischemia-reperfusion event in the central nervous system by (i) pro-inflammatory activation of astrocytes via ephrin-B-dependent signaling and (ii) amplification of NMDA-evoked neuronal excitotoxicity.


Subject(s)
Brain Ischemia/metabolism , Brain/metabolism , Encephalitis/metabolism , Neurons/metabolism , Receptor, EphB2/metabolism , Stroke/metabolism , Animals , Astrocytes/metabolism , Brain/pathology , Brain Ischemia/complications , Brain Ischemia/pathology , Encephalitis/complications , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Neurons/pathology , Receptor, EphB2/genetics , Signal Transduction , Stroke/complications , Stroke/pathology
11.
Mol Pain ; 15: 1744806919827469, 2019.
Article in English | MEDLINE | ID: mdl-30638145

ABSTRACT

Chronic pain is a pathological manifestation of neuronal plasticity supported by altered gene transcription in spinal cord neurons that results in long-lasting hypersensitivity. Recently, the concept that epigenetic regulators might be important in pathological pain has emerged, but a clear understanding of the molecular players involved in the process is still lacking. In this study, we linked Dnmt3a2, a synaptic activity-regulated de novo DNA methyltransferase, to chronic inflammatory pain. We observed that Dnmt3a2 levels are increased in the spinal cord of adult mice following plantar injection of Complete Freund's Adjuvant, an in vivo model of chronic inflammatory pain. In vivo knockdown of Dnmt3a2 expression in dorsal horn neurons blunted the induction of genes triggered by Complete Freund's Adjuvant injection. Among the genes whose transcription was found to be influenced by Dnmt3a2 expression in the spinal cord is Ptgs2, encoding for Cox-2, a prime mediator of pain processing. Lowering the levels of Dnmt3a2 prevented the establishment of long-lasting inflammatory hypersensitivity. These results identify Dnmt3a2 as an important epigenetic regulator needed for the establishment of central sensitization. Targeting expression or function of Dnmt3a2 may be suitable for the treatment of chronic pain.


Subject(s)
Chronic Pain/complications , DNA (Cytosine-5-)-Methyltransferases/metabolism , Epigenesis, Genetic , Hyperalgesia/metabolism , Inflammation/complications , Posterior Horn Cells/metabolism , Up-Regulation/physiology , Animals , Capsaicin/pharmacology , Cells, Cultured , Chronic Pain/chemically induced , Chronic Pain/pathology , Cyclooxygenase 1/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Disease Models, Animal , Escherichia coli Proteins/metabolism , Freund's Adjuvant/toxicity , Functional Laterality , Male , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Pain Measurement , Phosphopyruvate Hydratase/metabolism , Posterior Horn Cells/drug effects , Potassium Chloride/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Spinal Cord/pathology , Up-Regulation/drug effects
12.
Cereb Cortex ; 29(6): 2701-2715, 2019 06 01.
Article in English | MEDLINE | ID: mdl-29982364

ABSTRACT

Excessive excitation has been hypothesized to subsume a significant part of the acute damage occurring after traumatic brain injury (TBI). However, reduced neuronal excitability, loss of neuronal firing, and a disturbed excitation/inhibition balance have been detected. Parvalbumin (PV) interneurons are major regulators of perisomatic inhibition, principal neurons firing, and overall cortical excitability. However, their role in acute TBI pathogenic cascades is unclear. We exploited the chemogenetic Pharmacologically Selective Activation Module and Pharmacologically Selective Effector Module control of PV-Cre+ neurons and the Designer Receptors Exclusively Activated by Designer Drug (DREADD) control of principal neurons in a blunt model of TBI to explore the role of inhibition in shaping neuronal vulnerability to TBI. We demonstrated that inactivation of PV interneurons at the instance or soon after trauma enhances survival of principal neurons and reduces gliosis at 7 dpi whereas, activation of PV interneurons decreased neuronal survival. The protective effect of PV inactivation was suppressed by expressing the nuclear calcium buffer PV-nuclear localisation sequence in principal neurons, implying an activity-dependent neuroprotective signal. In fact, protective effects were obtained by increasing the excitability of principal neurons directly using DREADDs. Thus, we show that sustaining neuronal excitation in the early phases of TBI may reduce neuronal vulnerability by increasing activity-dependent survival, while excess activation of perisomatic inhibition is detrimental to neuronal integrity.


Subject(s)
Brain Injuries, Traumatic/physiopathology , Interneurons/physiology , Animals , Mice , Neurons/physiology , Parvalbumins/metabolism
13.
J Biol Chem ; 290(38): 23039-49, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26231212

ABSTRACT

Calcium-binding proteins (CaBPs) such as parvalbumin are part of the cellular calcium buffering system that determines intracellular calcium diffusion and influences the spatiotemporal dynamics of calcium signals. In neurons, CaBPs are primarily localized to the cytosol and function, for example, in nerve terminals in short-term synaptic plasticity. However, CaBPs are also expressed in the cell nucleus, suggesting that they modulate nuclear calcium signals, which are key regulators of neuronal gene expression. Here we show that the calcium buffering capacity of the cell nucleus in mouse hippocampal neurons regulates neuronal architecture by modulating the expression levels of VEGFD and the complement factor C1q-c, two nuclear calcium-regulated genes that control dendrite geometry and spine density, respectively. Increasing the levels of nuclear calcium buffers by means of expression of a nuclearly targeted form of parvalbumin fused to mCherry (PV.NLS-mC) led to a reduction in VEGFD expression and, as a result, to a decrease in total dendritic length and complexity. In contrast, mRNA levels of the synapse pruning factor C1q-c were increased in neurons expressing PV.NLS-mC, causing a reduction in the density and size of dendritic spines. Our results establish a close link between nuclear calcium buffering capacity and the transcription of genes that determine neuronal structure. They suggest that the development of cognitive deficits observed in neurological conditions associated with CaBP deregulation may reflect the loss of necessary structural features of dendrites and spines.


Subject(s)
Calcium/metabolism , Dendritic Spines/metabolism , Hippocampus/metabolism , Nerve Net/metabolism , Animals , Complement C1q/biosynthesis , Gene Expression Regulation/physiology , Hippocampus/cytology , Mice , Nerve Net/cytology , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor D/biosynthesis
14.
J Neurosurg Anesthesiol ; 27(3): 187-93, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25144502

ABSTRACT

BACKGROUND: Disturbed brain metabolism is a signature of primary damage and/or precipitates secondary injury processes after severe brain injury. Sedatives and analgesics target electrophysiological functioning and are as such well-known modulators of brain energy metabolism. Still unclear, however, is how sedatives impact glucose metabolism and whether they differentially influence brain metabolism in normally active, healthy brain and critically impaired, injured brain. We therefore examined and compared the effects of anesthetic drugs under both critical (<1 mmol/L) and noncritical (>1 mmol/L) extracellular brain glucose levels. METHODS: We performed an explorative, retrospective analysis of anesthetic drug administration and brain glucose concentrations, obtained by bedside microdialysis, in 19 brain-injured patients. RESULT: Our investigations revealed an inverse linear correlation between brain glucose and both the concentration of extracellular glutamate (Pearson r=-0.58, P=0.01) and the lactate/glucose ratio (Pearson r=-0.55, P=0.01). For noncritical brain glucose levels, we observed a positive linear correlation between midazolam dose and brain glucose (P<0.05). For critical brain glucose levels, extracellular brain glucose was unaffected by any type of sedative. CONCLUSIONS: These findings suggest that the use of anesthetic drugs may be of limited value in attempts to influence brain glucose metabolism in injured brain tissue.


Subject(s)
Brain Injuries/metabolism , Brain/drug effects , Brain/metabolism , Glucose/metabolism , Hypnotics and Sedatives/pharmacology , Microdialysis/methods , Adult , Aged , Dose-Response Relationship, Drug , Female , Glutamic Acid/metabolism , Humans , Lactic Acid/metabolism , Male , Midazolam/pharmacology , Middle Aged , Retrospective Studies
15.
Cell Tissue Res ; 357(2): 407-26, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25012522

ABSTRACT

Acute physiological pain, the unpleasant sensory response to a noxious stimulus, is essential for animals and humans to avoid potential injury. Pathological pain that persists after the original insult or injury has subsided, however, not only results in individual suffering but also imposes a significant cost on society. Improving treatments for long-lasting pathological pain requires a comprehensive understanding of the biological mechanisms underlying pain perception and the development of pain chronicity. In this review, we aim to highlight some of the major findings related to the involvement of neuronal calcium signaling in the processes that mediate chronic pain.


Subject(s)
Calcium Signaling , Chronic Pain/metabolism , Neurons/pathology , Animals , Chronic Disease , Chronic Pain/physiopathology , Diabetic Neuropathies/metabolism , Diabetic Neuropathies/physiopathology , Humans , Neuralgia/metabolism , Neuralgia/physiopathology , Neuronal Plasticity , Neurons/metabolism
16.
Nat Commun ; 4: 2034, 2013.
Article in English | MEDLINE | ID: mdl-23774321

ABSTRACT

The recent identification of the mitochondrial Ca(2+) uniporter gene (Mcu/Ccdc109a) has enabled us to address its role, and that of mitochondrial Ca(2+) uptake, in neuronal excitotoxicity. Here we show that exogenously expressed Mcu is mitochondrially localized and increases mitochondrial Ca(2+) levels following NMDA receptor activation, leading to increased mitochondrial membrane depolarization and excitotoxic cell death. Knockdown of endogenous Mcu expression reduces NMDA-induced increases in mitochondrial Ca(2+), resulting in lower levels of mitochondrial depolarization and resistance to excitotoxicity. Mcu is subject to dynamic regulation as part of an activity-dependent adaptive mechanism that limits mitochondrial Ca(2+) overload when cytoplasmic Ca(2+) levels are high. Specifically, synaptic activity transcriptionally represses Mcu, via a mechanism involving the nuclear Ca(2+) and CaM kinase-mediated induction of Npas4, resulting in the inhibition of NMDA receptor-induced mitochondrial Ca(2+) uptake and preventing excitotoxic death. This establishes Mcu and the pathways regulating its expression as important determinants of excitotoxicity, which may represent therapeutic targets for excitotoxic disorders.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling/drug effects , Cell Nucleus/metabolism , Neuroprotective Agents/pharmacology , Neurotoxins/toxicity , Repressor Proteins/metabolism , Transcription, Genetic/drug effects , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biological Transport/drug effects , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Death/drug effects , Cell Nucleus/drug effects , Cell Nucleus/enzymology , Gene Knockdown Techniques , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , RNA, Small Interfering/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/drug effects , Synapses/metabolism
17.
Neuron ; 77(1): 43-57, 2013 Jan 09.
Article in English | MEDLINE | ID: mdl-23312515

ABSTRACT

Persistent pain induced by noxious stimuli is characterized by the transition from normosensitivity to hypersensitivity. Underlying mechanisms are not well understood, although gene expression is considered important. Here, we show that persistent nociceptive-like activity triggers calcium transients in neuronal nuclei within the superficial spinal dorsal horn, and that nuclear calcium is necessary for the development of long-term inflammatory hypersensitivity. Using a nucleus-specific calcium signal perturbation strategy in vivo complemented by gene profiling, bioinformatics, and functional analyses, we discovered a pain-associated, nuclear calcium-regulated gene program in spinal excitatory neurons. This includes C1q, a modulator of synaptic spine morphogenesis, which we found to contribute to activity-dependent spine remodelling on spinal neurons in a manner functionally associated with inflammatory hypersensitivity. Thus, nuclear calcium integrates synapse-to-nucleus communication following noxious stimulation and controls a spinal genomic response that mediates the transition between acute and long-term nociceptive sensitization by modulating functional and structural plasticity.


Subject(s)
Calcium Signaling/physiology , Cell Nucleus/physiology , Chronic Pain/genetics , Genomics , Posterior Horn Cells/physiology , Animals , Animals, Newborn , Cells, Cultured , Chronic Pain/pathology , Genomics/methods , Inflammation/genetics , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/pathology , Neurons/physiology , Pain Measurement/methods , Posterior Horn Cells/pathology , Spinal Cord/cytology , Spinal Cord/pathology , Spinal Cord/physiology
18.
Neuroimage ; 60(1): 139-52, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22209812

ABSTRACT

Cognitive and behavioral functions depend on the activation of stable neuronal assemblies, i.e. distributed groups of co-active neurons within neuronal networks. It is therefore crucial to monitor distributed patterns of activity in real time with single-neuron resolution. Microelectrode recordings allow detection of coincidence between discharges of identified units at high temporal resolution, but are not able to reveal the full spatial pattern of activity in multi-cellular assemblies. Therefore, observation of such distributed sets of neurons is a stronghold of optical techniques, but the required resolution, sensitivity, and speed are still challenging current technology. Here, we report a new approach for monitoring neuronal assemblies, using memory-related network oscillations in rodent hippocampal circuits as a model. The cytosolic calcium-sensitive fluorescent protein GCaMP3.NES was expressed using recombinant adeno-associated viral (rAAV)-mediated gene transfer in CA3 pyramidal neurons of cultured mouse hippocampal slices. After 14-21 days in culture, field potential recordings revealed spontaneous occurrence of sharp wave-ripple network events during which a fraction of local neurons is coherently activated. Using a custom-built epi-fluorescence microscope we could monitor a field of view of 410 µm × 410 µm with single-neuron optical resolution (20× objective, 0.4 NA). We developed a highly sensitive and specific wavelet-based method of cell identification allowing simultaneous observation of more than 150 neurons at frame rates of up to 60 Hz. Our recording configuration and image analysis provide a tool to investigate cognition-related activity patterns in the hippocampus and other circuits.


Subject(s)
Hippocampus/cytology , Hippocampus/physiology , Neurons/physiology , Animals , Mice , Mice, Inbred C57BL , Neuroimaging , Reproducibility of Results , Tissue Culture Techniques
19.
Cold Spring Harb Perspect Biol ; 3(11): a004564, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21791697

ABSTRACT

Changes in the intracellular concentration of calcium ions in neurons are involved in neurite growth, development, and remodeling, regulation of neuronal excitability, increases and decreases in the strength of synaptic connections, and the activation of survival and programmed cell death pathways. An important aspect of the signals that trigger these processes is that they are frequently initiated in the form of glutamatergic neurotransmission within dendritic trees, while their completion involves specific changes in the patterns of genes expressed within neuronal nuclei. Accordingly, two prominent aims of research concerned with calcium signaling in neurons are determination of the mechanisms governing information conveyance between synapse and nucleus, and discovery of the rules dictating translation of specific patterns of inputs into appropriate and specific transcriptional responses. In this article, we present an overview of the avenues by which glutamatergic excitation of dendrites may be communicated to the neuronal nucleus and the primary calcium-dependent signaling pathways by which synaptic activity can invoke changes in neuronal gene expression programs.


Subject(s)
Calcium Signaling , Calcium/chemistry , Cell Nucleus/chemistry , Neurons/chemistry , Synapses/chemistry , Calcium/metabolism , Cell Nucleus/metabolism , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/physiology , Epigenesis, Genetic , Gene Expression Regulation , Models, Biological , Models, Genetic , Neurons/metabolism , Neurons/physiology , Nuclear Pore/chemistry , Nuclear Pore/physiology , Permeability , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/physiology , Synapses/metabolism
20.
J Physiol ; 589(Pt 13): 3211-29, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21576272

ABSTRACT

Group I metabotropic glutamate receptors (mGluRs) play an essential role in cognitive function. Their activation results in a wide array of cellular and molecular responses that are mediated by multiple signalling cascades. In this study, we focused on Group I mGluR activation of IP3R-mediated intracellular Ca2+ waves and their role in activating Ca2+-dependent ion channels in CA1 pyramidal neurons. Using whole-cell patch-clamp recordings and high-speed Ca2+ fluorescence imaging in acute hippocampal brain slices, we show that synaptic and pharmacological stimulation of mGluRs triggers intracellular Ca2+ waves and a biphasic electrical response composed of a transient Ca2+-dependent SK channel-mediated hyperpolarization and a TRPC-mediated sustained depolarization. The generation and magnitude of the SK channel-mediated hyperpolarization depended solely on the rise in intracellular Ca2+ concentration ([Ca2+]i), whereas the TRPC channel-mediated depolarization required both a small rise in [Ca2+]i and mGluR activation. Furthermore, the TRPC-mediated current was suppressed by forskolin-induced rises in cAMP. We also show that SK- and TRPC-mediated currents robustly modulate pyramidal neuron excitability by decreasing and increasing their firing frequency, respectively. These findings provide additional evidence that mGluR-mediated synaptic transmission makes an important contribution to regulating the output of hippocampal neurons through intracellular Ca2+ wave activation of SK and TRPC channels. cAMP provides an additional level of regulation by modulating TRPC-mediated sustained depolarization that we propose to be important for stabilizing periods of sustained firing.


Subject(s)
CA1 Region, Hippocampal/physiology , Calcium Signaling/physiology , Neurons/physiology , Pyramidal Cells/physiology , Receptors, Metabotropic Glutamate/physiology , Small-Conductance Calcium-Activated Potassium Channels/physiology , TRPC Cation Channels/physiology , Action Potentials/physiology , Animals , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...