Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Cell ; 187(15): 4043-4060.e30, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38878778

ABSTRACT

Inflammation-induced neurodegeneration is a defining feature of multiple sclerosis (MS), yet the underlying mechanisms remain unclear. By dissecting the neuronal inflammatory stress response, we discovered that neurons in MS and its mouse model induce the stimulator of interferon genes (STING). However, activation of neuronal STING requires its detachment from the stromal interaction molecule 1 (STIM1), a process triggered by glutamate excitotoxicity. This detachment initiates non-canonical STING signaling, which leads to autophagic degradation of glutathione peroxidase 4 (GPX4), essential for neuronal redox homeostasis and thereby inducing ferroptosis. Both genetic and pharmacological interventions that target STING in neurons protect against inflammation-induced neurodegeneration. Our findings position STING as a central regulator of the detrimental neuronal inflammatory stress response, integrating inflammation with glutamate signaling to cause neuronal cell death, and present it as a tractable target for treating neurodegeneration in MS.


Subject(s)
Inflammation , Membrane Proteins , Multiple Sclerosis , Neurons , Animals , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Membrane Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Mice , Humans , Inflammation/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Signal Transduction , Autophagy , Mice, Inbred C57BL , Glutamic Acid/metabolism , Ferroptosis , Disease Models, Animal , Female , Male
2.
J Clin Invest ; 134(16)2024 Jun 18.
Article in English | MEDLINE | ID: mdl-39145444

ABSTRACT

A disturbed balance between excitation and inhibition (E/I balance) is increasingly recognized as a key driver of neurodegeneration in multiple sclerosis (MS), a chronic inflammatory disease of the central nervous system. To understand how chronic hyperexcitability contributes to neuronal loss in MS, we transcriptionally profiled neurons from mice lacking inhibitory metabotropic glutamate signaling with shifted E/I balance and increased vulnerability to inflammation-induced neurodegeneration. This revealed a prominent induction of the nuclear receptor NR4A2 in neurons. Mechanistically, NR4A2 increased susceptibility to excitotoxicity by stimulating continuous VGF secretion leading to glycolysis-dependent neuronal cell death. Extending these findings to people with MS (pwMS), we observed increased VGF levels in serum and brain biopsies. Notably, neuron-specific deletion of Vgf in a mouse model of MS ameliorated neurodegeneration. These findings underscore the detrimental effect of a persistent metabolic shift driven by excitatory activity as a fundamental mechanism in inflammation-induced neurodegeneration.


Subject(s)
Glycolysis , Inflammation , Neurons , Nuclear Receptor Subfamily 4, Group A, Member 2 , Animals , Mice , Humans , Neurons/metabolism , Neurons/pathology , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation/genetics , Multiple Sclerosis/pathology , Multiple Sclerosis/metabolism , Multiple Sclerosis/genetics , Mice, Knockout , Signal Transduction , Male , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology
3.
Sci Adv ; 9(47): eadi6855, 2023 11 24.
Article in English | MEDLINE | ID: mdl-38000031

ABSTRACT

Neuroinflammation causes neuronal injury in multiple sclerosis (MS) and other neurological diseases. MicroRNAs (miRNAs) are important modulators of neuronal stress responses, but knowledge about their contribution to neuronal protection or damage during inflammation is limited. Here, we constructed a regulatory miRNA-mRNA network of inflamed motor neurons by leveraging cell type-specific miRNA and mRNA sequencing of mice undergoing experimental autoimmune encephalomyelitis (EAE). We found robust induction of miR-92a in inflamed spinal cord neurons and identified cytoplasmic polyadenylation element-binding protein 3 (Cpeb3) as a key target of miR-92a-mediated posttranscriptional silencing. We detected CPEB3 repression in inflamed neurons in murine EAE and human MS. Moreover, both miR-92a delivery and Cpeb3 deletion protected neuronal cultures against excitotoxicity. Supporting a detrimental effect of Cpeb3 in vivo, neuron-specific deletion in conditional Cpeb3 knockout animals led to reduced inflammation-induced clinical disability in EAE. Together, we identified a neuroprotective miR-92a-Cpeb3 axis in neuroinflammation that might serve as potential treatment target to limit inflammation-induced neuronal damage.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , MicroRNAs , Multiple Sclerosis , Humans , Mice , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Neuroinflammatory Diseases , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Inflammation/genetics , Inflammation/metabolism , Neurons/metabolism , RNA, Messenger/metabolism , Mice, Inbred C57BL , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
4.
Sci Adv ; 8(31): eabm5500, 2022 08 05.
Article in English | MEDLINE | ID: mdl-35930635

ABSTRACT

Neuroinflammation leads to neuronal stress responses that contribute to neuronal dysfunction and loss. However, treatments that stabilize neurons and prevent their destruction are still lacking. Here, we identify the histone methyltransferase G9a as a druggable epigenetic regulator of neuronal vulnerability to inflammation. In murine experimental autoimmune encephalomyelitis (EAE) and human multiple sclerosis (MS), we found that the G9a-catalyzed repressive epigenetic mark H3K9me2 was robustly induced by neuroinflammation. G9a activity repressed anti-ferroptotic genes, diminished intracellular glutathione levels, and triggered the iron-dependent programmed cell death pathway ferroptosis. Conversely, pharmacological treatment of EAE mice with a G9a inhibitor restored anti-ferroptotic gene expression, reduced inflammation-induced neuronal loss, and improved clinical outcome. Similarly, neuronal anti-ferroptotic gene expression was reduced in MS brain tissue and was boosted by G9a inhibition in human neuronal cultures. This study identifies G9a as a critical transcriptional enhancer of neuronal ferroptosis and potential therapeutic target to counteract inflammation-induced neurodegeneration.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Ferroptosis , Multiple Sclerosis , Animals , Encephalomyelitis, Autoimmune, Experimental/genetics , Ferroptosis/genetics , Gene Expression Regulation , Histone-Lysine N-Methyltransferase/metabolism , Humans , Inflammation/genetics , Mice , Neurons/metabolism
5.
J Exp Med ; 218(5)2021 05 03.
Article in English | MEDLINE | ID: mdl-33661276

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system with continuous neuronal loss. Treatment of clinical progression remains challenging due to lack of insights into inflammation-induced neurodegenerative pathways. Here, we show that an imbalance in the neuronal receptor interactome is driving glutamate excitotoxicity in neurons of MS patients and identify the MS risk-associated metabotropic glutamate receptor 8 (GRM8) as a decisive modulator. Mechanistically, GRM8 activation counteracted neuronal cAMP accumulation, thereby directly desensitizing the inositol 1,4,5-trisphosphate receptor (IP3R). This profoundly limited glutamate-induced calcium release from the endoplasmic reticulum and subsequent cell death. Notably, we found Grm8-deficient neurons to be more prone to glutamate excitotoxicity, whereas pharmacological activation of GRM8 augmented neuroprotection in mouse and human neurons as well as in a preclinical mouse model of MS. Thus, we demonstrate that GRM8 conveys neuronal resilience to CNS inflammation and is a promising neuroprotective target with broad therapeutic implications.


Subject(s)
Central Nervous System/metabolism , Inflammation/genetics , Neurodegenerative Diseases/genetics , Receptors, Metabotropic Glutamate/genetics , Animals , Cell Survival/genetics , Cells, Cultured , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Gene Expression Profiling/methods , Gene Regulatory Networks/genetics , Humans , Inflammation/metabolism , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Neurodegenerative Diseases/metabolism , Neurons/cytology , Neurons/metabolism , Neuroprotective Agents/metabolism , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction/genetics
6.
Nat Neurosci ; 22(6): 887-896, 2019 06.
Article in English | MEDLINE | ID: mdl-31011226

ABSTRACT

Multiple sclerosis (MS) is characterized by inflammatory insults that drive neuroaxonal injury. However, knowledge about neuron-intrinsic responses to inflammation is limited. By leveraging neuron-specific messenger RNA profiling, we found that neuroinflammation leads to induction and toxic accumulation of the synaptic protein bassoon (Bsn) in the neuronal somata of mice and patients with MS. Neuronal overexpression of Bsn in flies resulted in reduction of lifespan, while genetic disruption of Bsn protected mice from inflammation-induced neuroaxonal injury. Notably, pharmacological proteasome activation boosted the clearance of accumulated Bsn and enhanced neuronal survival. Our study demonstrates that neuroinflammation initiates toxic protein accumulation in neuronal somata and advocates proteasome activation as a potential remedy.


Subject(s)
Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Nerve Tissue Proteins/metabolism , Animals , Drosophila , Humans , Inflammation/metabolism , Inflammation/pathology , Mice , Neurons/metabolism , Neurons/pathology , Spinal Cord/metabolism , Spinal Cord/pathology
7.
Cell Rep ; 23(9): 2705-2717, 2018 05 29.
Article in English | MEDLINE | ID: mdl-29847800

ABSTRACT

Autism spectrum disorders (ASDs) are associated with mutations affecting synaptic components, including GluN2B-NMDA receptors (NMDARs) and neurobeachin (NBEA). NBEA participates in biosynthetic pathways to regulate synapse receptor targeting, synaptic function, cognition, and social behavior. However, the role of NBEA-mediated transport in specific trafficking routes is unclear. Here, we highlight an additional function for NBEA in the local delivery and surface re-insertion of synaptic receptors in mouse neurons. NBEA dynamically interacts with Rab4-positive recycling endosomes, transiently enters spines in an activity-dependent manner, and regulates GluN2B-NMDAR recycling. Furthermore, we show that the microtubule growth inhibitor kinesin KIF21B constrains NBEA dynamics and is present in the NBEA-recycling endosome-NMDAR complex. Notably, Kif21b knockout decreases NMDAR surface expression and alters social behavior in mice, consistent with reported social deficits in Nbea mutants. The influence of NBEA-KIF21B interactions on GluN2B-NMDAR local recycling may be relevant to mechanisms underlying ASD etiology.


Subject(s)
Behavior, Animal , Carrier Proteins/metabolism , Endocytosis , Kinesins/metabolism , Nerve Tissue Proteins/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Social Behavior , Animals , COS Cells , Chlorocebus aethiops , Cognition , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Dyneins/metabolism , Endocytosis/drug effects , Endosomes/metabolism , Glutamic Acid/pharmacology , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Membrane Proteins , Mice, Knockout , Microtubules/drug effects , Microtubules/metabolism , Nocodazole/pharmacology , Protein Binding/drug effects , Protein Transport/drug effects , Synaptic Vesicles/drug effects , Synaptic Vesicles/metabolism , rab4 GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL