1
|
Fletcher R, Hoppe M, McQuail JA, Hernandez CM, Hernandez AR. Ketogenic Diet-Induced Alterations in Neuronal Signaling-Related Proteins are Not Due to Differences in Synaptosome Protein Levels. Mol Neurobiol 2025:10.1007/s12035-025-04988-1. [PMID: 40299298 DOI: 10.1007/s12035-025-04988-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 04/18/2025] [Indexed: 04/30/2025]
Abstract
Impaired cognitive function is a hallmark of advancing age, and the potential to reverse or delay these cognitive deficits through dietary intervention holds promise for improving quality of life for older adults. Specifically, ketogenic diets (KDs) have now been widely used for the treatment of several neurological and peripheral disorders, including diseases profoundly affecting cognitive health, of which advanced age is the single greatest risk factor. However, the precise mechanisms of the efficacy of KD-based interventions to reverse age-related cognitive and neurobiological declines are not fully elucidated. We have previously demonstrated that a KD improves cognitive function, with concurrent increases in global levels of proteins related to synaptic signaling in the aging hippocampus (HPC) and prefrontal cortex (PFC). Despite these advances, it remains unclear as to whether these changes reflect biochemical modifications specifically localized to synaptic terminals. To address this important, unanswered question, we purified synaptosomes from the HPC and PFC of aging rats fed a KD or control diet (CD) for a minimum of 4 months and quantified 10 proteins related to synaptic transmission. In contrast to previous studies of global protein expression, the signaling proteins measured did not show significant differences between diet groups in synaptosomes isolated from either region. When pre-mortem performance on an Object-Place Paired Association task was considered, we found a significant correlation between several proteins within the HPC and PFC synaptosomes of CD-fed rats, more pronounced in CD-fed aged rats, that are conspicuously absent in KD-fed rats from both age groups. Moreover, there is a significant alteration in the ratio of VGAT/VGluT1, markers of excitatory and inhibitory synaptic vesicles, in the PFC with dietary intervention that is absent in the HPC, confirming prior reports of regionally specific alterations in excitatory and inhibitory signaling post KD. These new and extended findings reveal important, naturally occurring associations between protein levels localized to synaptic terminals, while clarifying that effects KD likely increase synaptic abundance without altering the biochemical composition of isolated synapses.
Collapse
Affiliation(s)
| | - Meagan Hoppe
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Joseph A McQuail
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Caesar M Hernandez
- Department of Medicine, Division of Geriatrics, University of Alabama at Birmingham, Gerontology & Palliative Care, 845 19th St. South Rm 768, Birmingham, AL, 35205, USA
| | - Abbi R Hernandez
- Department of Medicine, Division of Geriatrics, University of Alabama at Birmingham, Gerontology & Palliative Care, 845 19th St. South Rm 768, Birmingham, AL, 35205, USA.
| |
Collapse
|
2
|
Yuan C, Patel K, Shi H, Wang HLV, Wang F, Li R, Li Y, Corces VG, Shi H, Das S, Yu J, Jin P, Yao B, Hu J. mcDETECT: Decoding 3D Spatial Synaptic Transcriptomes with Subcellular-Resolution Spatial Transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.27.645744. [PMID: 40236251 PMCID: PMC11996425 DOI: 10.1101/2025.03.27.645744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Spatial transcriptomics (ST) has shown great potential for unraveling the molecular mechanisms of neurodegenerative diseases. However, most existing analyses of ST data focus on bulk or single-cell resolution, overlooking subcellular compartments such as synapses, which are fundamental structures of the brain's neural network. Here we present mcDETECT, a novel framework that integrates machine learning algorithms and in situ ST (iST) with targeted gene panels to study synapses. mcDETECT identifies individual synapses based on the aggregation of synaptic mRNAs in three-dimensional (3D) space, allowing for the construction of single-synapse spatial transcriptome profiles. By benchmarking the synapse density measured by volume electron microscopy and genetic labeling, we demonstrate that mcDETECT can faithfully and accurately recover the spatial location of single synapses using iST data from multiple platforms, including Xenium, Xenium 5K, MERSCOPE, and CosMx. Based on the subsequent transcriptome profiling, we further stratify total synapses into various subtypes and explore their pathogenic dysregulation associated with Alzheimer's disease (AD) progression, which provides potential targets for synapse-specific therapies in AD progression.
Collapse
|
3
|
Yasmin F, Marwick KFM, Hunter DW, Nawaz S, Marshall GF, Booker SA, Hardingham GE, Kind PC, Wyllie DJA. Absence of GluN2A in hippocampal CA1 neurons leads to altered dendritic structure and reduced frequency of miniature excitatory synaptic events. Brain Commun 2025; 7:fcaf124. [PMID: 40226380 PMCID: PMC11986202 DOI: 10.1093/braincomms/fcaf124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 02/04/2025] [Accepted: 03/25/2025] [Indexed: 04/15/2025] Open
Abstract
GluN2A is a NMDA receptor subunit postulated as important for learning and memory. In humans, heterozygous loss of function variants in the gene encoding it (GRIN2A) increase the risk of epilepsy, intellectual disability and schizophrenia. Haploinsufficient mouse models show electrophysiological abnormalities and thus to improve and widen understanding of the pathogenesis of GRIN2A-associated disorders in humans, this study aimed to assess the impact of Grin2a absence and haploinsufficiency on core neuronal and synaptic properties in genetically modified rats. Electrophysiological whole-cell current- and voltage-clamp recordings were made from CA1 pyramidal neurons in acute hippocampal slices from wild-type and Grin2a heterozygous (Grin2a+/- ) and homozygous (Grin2a-/- ) knock out rats aged postnatal day 27-34. While reduced levels or absence of GluN2A did not affect neuronal excitability or intrinsic membrane properties in both Grin2a+/- and Grin2a-/- rats, we found a reduced frequency of miniature excitatory post synaptic currents and a reduced density of proximal dendrites suggestive of a reduced number of excitatory synapses. Recordings from CA1 neurons in slices prepared from Grin2a+/- and Grin2a-/- rats revealed there was a reduced ratio of the current mediated by NMDA receptors compared to AMPA receptors, while in Grin2a-/- recordings, there was a slowing of the decay time-constant of the NMDA receptor-mediated excitatory postsynaptic currents. Moreover, neither summation of sub-threshold excitatory postsynaptic potentials nor summation of supra-threshold excitatory postsynaptic potentials to initiate action potential firing in CA1 pyramidal neurons indicated any dependence on GluN2A. We conclude that reduced levels of GluN2A alters the kinetics of NMDA receptor-mediated synaptic events and dendritic structure of CA1 neurons, but do not affect several other core neuronal functions. These relatively subtle changes are consistent with the largely intact neural functioning of the majority of humans carrying GRIN2A loss of function variants. Further research could explore whether the changes in synaptic properties we observed contribute to alterations in higher level circuit dynamics and computation, which may manifest as disorders of cognition and excitability in humans.
Collapse
Affiliation(s)
- Farhana Yasmin
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Katie F M Marwick
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Daniel W Hunter
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Sarfaraz Nawaz
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Grant F Marshall
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Sam A Booker
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Giles E Hardingham
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, Edinburgh EH16 4SB, UK
| | - Peter C Kind
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - David J A Wyllie
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| |
Collapse
|
4
|
Chik MW, Meor Mohd Affandi MMR, Mohd Nor Hazalin NA, Surindar Singh GK. Astaxanthin nanoemulsion improves cognitive function and synaptic integrity in Streptozotocin-induced Alzheimer's disease model. Metab Brain Dis 2025; 40:136. [PMID: 40047916 DOI: 10.1007/s11011-025-01560-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 02/22/2025] [Indexed: 03/26/2025]
Abstract
Astaxanthin derived from natural sources has excellent antioxidant and anti-inflammatory effects, and it is currently being widely researched as a neuroprotectant. However, astaxanthin possesses low oral bioavailability, and thus, astaxanthin extract from Haematococcus pluvialis was formulated into a nanoemulsion to improve its bioavailability and administered to Alzheimer's disease (AD)-like rats to study its possible neuroprotective benefits. Astaxanthin nanoemulsion was administered orally once a day for 28 days to streptozotocin (STZ)-induced AD rats at concentrations of 160, 320, and 640 mg/kg of body weight (bw) and subsequently assessed for cognitive function using behavioral assessments. Brain samples were collected for the assessment of AD biomarkers. Astaxanthin nanoemulsion at a dosage of 640 mg/kg bw significantly improved spatial learning, spatial memory, and recognition memory against STZ-AD rats. At 320 and 640 mg/kg bw, astaxanthin nanoemulsion significantly reduced levels of hippocampus synaptosomal amyloid beta and paired-helical fibrillary tau protein while increasing neuron survival. Additionally, astaxanthin nanoemulsion at 640 mg/kg bw significantly increased acetylcholine levels in the hippocampus and cerebellum. Astaxanthin nanoemulsion at all treatment dosages significantly reduced malondialdehyde, a lipid peroxidation product, and neuroinflammatory mediators (GFAP and TNF-α). Astaxanthin nanoemulsion supplementation has the potential to improve cognitive function and synaptic function by lowering amyloid beta and tau levels, as well as preserve neuron integrity by reducing neuroinflammation and lipid peroxidation, indicating that it may be able to treat some of the underlying causes of AD.
Collapse
Affiliation(s)
- Mazzura Wan Chik
- Department of Pharmacology and Life Sciences, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Selangor Branch, Puncak Alam Campus, Puncak Alam, 42300, Selangor, Malaysia
| | - Meor Mohd Redzuan Meor Mohd Affandi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Puncak Alam Campus, Puncak Alam, 42300, Selangor, Malaysia
| | - Nurul Aqmar Mohd Nor Hazalin
- Department of Pharmacology and Life Sciences, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Selangor Branch, Puncak Alam Campus, Puncak Alam, 42300, Selangor, Malaysia
- Integrative Pharmacogenomics Institute (iPROMiSE), Level 7, FF3, Universiti Teknologi MARA, Selangor Branch, Puncak Alam Campus, Bandar Puncak Alam, Selangor, 42300, Malaysia
| | - Gurmeet Kaur Surindar Singh
- Department of Pharmacology and Life Sciences, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Selangor Branch, Puncak Alam Campus, Puncak Alam, 42300, Selangor, Malaysia.
- Brain Degeneration and Therapeutics Group, Pharmaceutical and Life Sciences Community of Research, Universiti Teknologi MARA (UiTM), Shah Alam, Selangor, 40450, Malaysia.
| |
Collapse
|
5
|
Pushie MJ, Sylvain NJ, Hou H, Pendleton N, Wang R, Zimmermann L, Pally M, Cayabyab FS, Peeling L, Kelly ME. X-ray fluorescence mapping of brain tissue reveals the profound extent of trace element dysregulation in stroke pathophysiology. Metallomics 2024; 16:mfae054. [PMID: 39547935 PMCID: PMC11631071 DOI: 10.1093/mtomcs/mfae054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 11/13/2024] [Indexed: 11/17/2024]
Abstract
The brain is a privileged organ with regard to its trace element composition and maintains a robust barrier system to sequester this specialized environment from the rest of the body and the vascular system. Stroke is caused by loss of adequate blood flow to a region of the brain. Without adequate blood flow ischaemic changes begin almost immediately, triggering an ischaemic cascade, characterized by ion dysregulation, loss of function, oxidative damage, cellular degradation, and breakdown of the barrier that helps maintain this environment. Ion dysregulation is a hallmark of stroke pathophysiology and we observe that most elements in the brain are dysregulated after stroke. X-ray fluorescence-based detection of physiological changes in the neurometallome after stroke reveals profound ion dysregulation within the lesion and surrounding tissue. Not only are most elements significantly dysregulated after stroke, but the level of dysregulation cannot be predicted from a cell-level description of dysregulation. X-ray fluorescence imaging reveals that the stroke lesion retains <25% of essential K+ after stroke, but this element is not concomitantly elevated elsewhere in the organ. Moreover, elements like Na+, Ca2+, and Cl- are vastly elevated above levels available in normal brain tissue (>400%, >200%, and >150%, respectively). We hypothesize that weakening of the blood-brain barrier after stroke allows elements to freely diffuse down their concentration gradient so that the stroke lesion is in equilibrium with blood (and the compartments containing brain interstitial fluid and cerebrospinal fluid). The change observed for the neurometallome likely has consequences for the potential to rescue infarcted tissue, but also presents specific targets for treatment.
Collapse
Affiliation(s)
- M Jake Pushie
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Nicole J Sylvain
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Huishu Hou
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Nicole Pendleton
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Richard Wang
- College of Medicine, Department of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Liam Zimmermann
- College of Medicine, Department of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Maxwell Pally
- College of Arts & Science, Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Francisco S Cayabyab
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Lissa Peeling
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Michael E Kelly
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| |
Collapse
|
6
|
Shaw GA, Wegener AJ, Neigh GN. Chronic corticosterone administration alters synaptic mitochondrial function within the hippocampus of C57Bl/6NTac mice. Physiol Behav 2024; 287:114681. [PMID: 39209050 PMCID: PMC12021453 DOI: 10.1016/j.physbeh.2024.114681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 08/02/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Chronic activation of the hypothalamic-pituitary-adrenal axis increases circulating corticosterone levels, causing a host of downstream behavioral, molecular, and metabolic changes. Here, we assess the effects of chronic exogenous CORT administration on changes in behavior and mitochondrial respiration in hippocampal synaptosomes of male and female mice. Adult male (n = 15) and female (n = 17) C57Bl/6NTac mice were given 35ug/mL CORT or vehicle dissolved in their drinking water for 21 consecutive days. Chronic CORT increased piloerection in males only. Although volume of CORT-containing water consumed was similar between males and females, circulating plasma and fecal corticosterone levels were only elevated in CORT-exposed males. Behavioral effects of CORT were evident in the Y-maze such that CORT caused a decrease in direct revisits in both sexes. There was no observed presentation of anxiety-like behavior following chronic CORT administration. Functional hippocampal synaptosomes were analyzed for mitochondrial respiration using Agilent's Cell Mito Stress test. Chronic CORT caused a decrease in synaptic mitochondria basal respiration, maximal respiration, proton leak, and ATP production in both sexes. Despite only observing an effect of chronic CORT on corticosterone concentrations in fecal and blood samples of males, chronic CORT induced marked changes in hippocampal synaptic mitochondrial function of both sexes. These data highlight the importance of considering effects of stress hormone exposure on neural function even in the absence of measurable peripheral elevations in females.
Collapse
Affiliation(s)
- Gladys A Shaw
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - Amy J Wegener
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - Gretchen N Neigh
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
7
|
Baytas O, Davidson SM, Kauer JA, Morrow EM. Loss of mitochondrial enzyme GPT2 leads to reprogramming of synaptic glutamate metabolism. Mol Brain 2024; 17:87. [PMID: 39604975 PMCID: PMC11600823 DOI: 10.1186/s13041-024-01154-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024] Open
Abstract
Recessive loss-of-function mutations in the mitochondrial enzyme Glutamate Pyruvate Transaminase 2 (GPT2) cause intellectual disability in children. Given this cognitive disorder, and because glutamate metabolism is tightly regulated to sustain excitatory neurotransmission, here we investigate the role of GPT2 in synaptic function. GPT2 catalyzes a reversible reaction interconverting glutamate and pyruvate with alanine and alpha-ketoglutarate, a TCA cycle intermediate; thereby, GPT2 may play an important role in linking mitochondrial tricarboxylic acid (TCA) cycle with synaptic transmission. In mouse brain, we find that GPT2 is enriched in mitochondria of synaptosomes (isolated synaptic terminals). Loss of Gpt2 in mouse appears to lead to reprogramming of glutamate and glutamine metabolism, and to decreased glutamatergic synaptic transmission. Whole-cell patch-clamp recordings in pyramidal neurons of CA1 hippocampal slices from Gpt2-null mice reveal decreased excitatory post-synaptic currents (mEPSCs) without changes in mEPSC frequency, or importantly, changes in inhibitory post-synaptic currents (mIPSCs). Additional evidence of defective glutamate release included reduced levels of glutamate released from Gpt2-null synaptosomes measured biochemically. Glutamate release from synaptosomes was rescued to wild-type levels by alpha-ketoglutarate supplementation. Additionally, we observed evidence of altered metabolism in isolated Gpt2-null synaptosomes: decreased TCA cycle intermediates, and increased glutamate dehydrogenase activity. Notably, alterations in the TCA cycle and the glutamine pool were alleviated by alpha-ketoglutarate supplementation. In conclusion, our data support a model whereby GPT2 mitochondrial activity may contribute to glutamate availability in pre-synaptic terminals, thereby highlighting potential interactions between pre-synaptic mitochondrial metabolism and synaptic transmission.
Collapse
Affiliation(s)
- Ozan Baytas
- Department of Molecular Biology, Cell Biology and Biochemistry, Laboratories for Molecular Medicine, Brown University, 70 Ship Street, Providence, RI, 02912, USA
- Center for Translational Neuroscience, Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, RI, 02912, USA
- Neuroscience Graduate Program, Brown University, Providence, RI, 02912, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Shawn M Davidson
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, 08540, USA
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Julie A Kauer
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94035, USA
| | - Eric M Morrow
- Department of Molecular Biology, Cell Biology and Biochemistry, Laboratories for Molecular Medicine, Brown University, 70 Ship Street, Providence, RI, 02912, USA.
- Center for Translational Neuroscience, Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, RI, 02912, USA.
| |
Collapse
|
8
|
Wang G, Peng S, Reyes Mendez M, Keramidas A, Castellano D, Wu K, Han W, Tian Q, Dong L, Li Y, Lu W. The TMEM132B-GABA A receptor complex controls alcohol actions in the brain. Cell 2024; 187:6649-6668.e35. [PMID: 39357522 DOI: 10.1016/j.cell.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 07/19/2024] [Accepted: 09/04/2024] [Indexed: 10/04/2024]
Abstract
Alcohol is the most consumed and abused psychoactive drug globally, but the molecular mechanisms driving alcohol action and its associated behaviors in the brain remain enigmatic. Here, we have discovered a transmembrane protein TMEM132B that is a GABAA receptor (GABAAR) auxiliary subunit. Functionally, TMEM132B promotes GABAAR expression at the cell surface, slows receptor deactivation, and enhances the allosteric effects of alcohol on the receptor. In TMEM132B knockout (KO) mice or TMEM132B I499A knockin (KI) mice in which the TMEM132B-GABAAR interaction is specifically abolished, GABAergic transmission is decreased and alcohol-induced potentiation of GABAAR-mediated currents is diminished in hippocampal neurons. Behaviorally, the anxiolytic and sedative/hypnotic effects of alcohol are markedly reduced, and compulsive, binge-like alcohol consumption is significantly increased. Taken together, these data reveal a GABAAR auxiliary subunit, identify the TMEM132B-GABAAR complex as a major alcohol target in the brain, and provide mechanistic insights into alcohol-related behaviors.
Collapse
Affiliation(s)
- Guohao Wang
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shixiao Peng
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Miriam Reyes Mendez
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Angelo Keramidas
- Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Road, St Lucia, Brisbane, QLD 4072, Australia
| | - David Castellano
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kunwei Wu
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wenyan Han
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Qingjun Tian
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lijin Dong
- Genetic Engineering Core, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yan Li
- Proteomics Core Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei Lu
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
9
|
Pinkston BTC, Browning JL, Olsen ML. Astrocyte TrkB.T1 deficiency disrupts glutamatergic synaptogenesis and astrocyte-synapse interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619696. [PMID: 39484608 PMCID: PMC11526899 DOI: 10.1101/2024.10.22.619696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Perisynaptic astrocyte processes (PAPs) contact pre- and post-synaptic elements to provide structural and functional support to synapses. Accumulating research demonstrates that the cradling of synapses by PAPs is critical for synapse formation, stabilization, and plasticity. The specific signaling pathways that govern these astrocyte-synapse interactions, however, remain to be elucidated. Herein, we demonstrate a role for the astrocyte TrkB.T1 receptor, a truncated isoform of the canonical receptor for brain derived neurotrophic factor (BDNF), in modulating astrocyte-synapse interactions and excitatory synapse development. Neuron-astrocyte co-culture studies revealed that loss of astrocyte TrkB.T1 disrupts the formation of PAPs. To elucidate the role of TrkB.T1 in synapse development, we conditionally deleted TrkB.T1 in astrocytes in mice. Synaptosome preparations were employed to probe for TrkB.T1 localization at the PAP, and confocal three-dimensional microscopy revealed a significant reduction in synapse density and astrocyte-synapse interactions across development in the absence of astrocytic TrkB.T1. These findings suggest that BDNF/TrkB.T1 signaling in astrocytes is critical for normal excitatory synapse formation in the cortex and that astrocyte TrkB.T1 serves a requisite role in astrocyte synapse interactions. Overall, this work provides new insights into the molecular mechanisms of astrocyte-mediated synaptogenesis and may have implications for understanding neurodevelopmental disorders and developing potential therapeutic targets.
Collapse
|
10
|
Asemi R, Ahmadi Asouri S, Aghadavod E, Jamilian M. The beneficial influences of vitamin D intake on inflammation and oxidative stress in infertile women with polycystic ovary syndrome. Ann Med Surg (Lond) 2024; 86:5218-5223. [PMID: 39239011 PMCID: PMC11374188 DOI: 10.1097/ms9.0000000000002349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 06/26/2024] [Indexed: 09/07/2024] Open
Abstract
Objective Oxidative stress and inflammation play a vital function in the pathophysiology of polycystic ovary syndrome (PCOS) and infertility. The aim of this work was to control the impacts of vitamin D intake on metabolic profiles in infertile subjects with PCOS. Trial design and methods This randomized, double-blinded, placebo-controlled clinical trial was carried out among 40 infertile women with PCOS. Subjects were randomly divided into two intervention groups to take either 50 000 IU vitamin D (n=20) or placebo (n=20) weekly for 8 weeks. Metabolic profiles and few inflammatory cytokines expression evaluated on peripheral blood mononuclear cells (PBMCs) of participants, using real-time polymerase chain reaction (RT-PCR) method. Results Vitamin D intake decreased high-sensitivity C-reactive protein (hs-CRP) (-0.9±1.1 vs. 0.3±0.9 mg/l, P=0.002) and elevated total antioxidant capacity (TAC) levels (49.2±60.2 vs. -50.6±161.8 mmol/l, P=0.02) compared with placebo; but no significant effects on other metabolic parameters were observed. Moreover, a significant downregulation of tumor necrosis factor alpha (TNF-α) expression (P=0.03) was observed after taking vitamin D compared with the placebo. Conclusions Overall, vitamin D intake for eight weeks had beneficial impacts on hs-CRP, TAC, and TNF-α among infertile women with PCOS.
Collapse
Affiliation(s)
- Reza Asemi
- Department of Internal Medicine, School of Medicine, Cancer Prevention Research Center, Seyyed Al-Shohada Hospital, Department of Internal Medicine, Isfahan University of Medical Sciences, Isfahan
| | - Sahar Ahmadi Asouri
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Department of Biochemistry, Kashan University of Medical Sciences, Kashan
| | - Esmat Aghadavod
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Department of Biochemistry, Kashan University of Medical Sciences, Kashan
| | - Mehri Jamilian
- Traditional and Complementary Medicine Research Center, Department of Gynecology, Arak University of Medical Sciences, Arak, Iran
| |
Collapse
|
11
|
Lu CW, Lin TY, Chang YY, Chiu KM, Lee MY, Wang SJ. Albiflorin Decreases Glutamate Release from Rat Cerebral Cortex Nerve Terminals (Synaptosomes) through Depressing P/Q-Type Calcium Channels and Protein Kinase A Activity. Int J Mol Sci 2024; 25:8846. [PMID: 39201534 PMCID: PMC11354331 DOI: 10.3390/ijms25168846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
The purpose of this study was to investigate whether and how albiflorin, a natural monoterpene glycoside, affects the release of glutamate, one of the most important neurotransmitters involved in neurotoxicity, from cerebrocortical nerve terminals (synaptosomes) in rats. The results showed that albiflorin reduced 4-aminopyridine (4-AP)-elicited glutamate release from synaptosomes, which was abrogated in the absence of extracellular Ca2+ or in the presence of the vesicular glutamate transporter inhibitor or a P/Q-type Ca2+ channel inhibitor, indicating a mechanism of action involving Ca2+-dependent depression of vesicular exocytotic glutamate release. Albiflorin failed to alter the increase in the fluorescence intensity of 3,3-diethylthiacarbocyanine iodide (DiSC3(5)), a membrane-potential-sensitive dye. In addition, the suppression of protein kinase A (PKA) abolished the effect of albiflorin on glutamate release. Albiflorin also reduced the phosphorylation of PKA and synaptosomal-associated protein of 25 kDa (SNAP-25) and synapsin I at PKA-specific residues, which correlated with decreased available synaptic vesicles. The results of transmission electron microscopy (TEM) also observed that albiflorin reduces the release competence of synaptic vesicles evoked by 4-AP in synaptosomes. In conclusion, by studying synaptosomally released glutamate, we suggested that albiflorin reduces vesicular exocytotic glutamate release by decreasing extracellular Ca2+ entry via P/Q-type Ca2+ channels and reducing PKA-mediated synapsin I and SNAP-25 phosphorylation.
Collapse
Affiliation(s)
- Cheng-Wei Lu
- Department of Anesthesiology, Far-Eastern Memorial Hospital, New Taipei City 22060, Taiwan; (C.-W.L.); (T.-Y.L.); (Y.-Y.C.)
- Department of Mechanical Engineering, Yuan Ze University, Taoyuan 32003, Taiwan
| | - Tzu-Yu Lin
- Department of Anesthesiology, Far-Eastern Memorial Hospital, New Taipei City 22060, Taiwan; (C.-W.L.); (T.-Y.L.); (Y.-Y.C.)
- Department of Mechanical Engineering, Yuan Ze University, Taoyuan 32003, Taiwan
| | - Ya-Ying Chang
- Department of Anesthesiology, Far-Eastern Memorial Hospital, New Taipei City 22060, Taiwan; (C.-W.L.); (T.-Y.L.); (Y.-Y.C.)
- International Program in Engineering for Bachelor, Yuan Ze University, Taoyuan 32003, Taiwan
| | - Kuan-Ming Chiu
- Division of Cardiovascular Surgery, Cardiovascular Center, Far-Eastern Memorial Hospital, New Taipei City 22060, Taiwan;
- Department of Electrical Engineering, Yuan Ze University, Taoyuan 32003, Taiwan
| | - Ming-Yi Lee
- Department of Medical Research, Far-Eastern Memorial Hospital, New Taipei City 22060, Taiwan;
| | - Su-Jane Wang
- School of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan
| |
Collapse
|
12
|
Zoltowska KM, Das U, Lismont S, Enzlein T, Maesako M, Houser MCQ, Franco ML, Özcan B, Gomes Moreira D, Karachentsev D, Becker A, Hopf C, Vilar M, Berezovska O, Mobley W, Chávez-Gutiérrez L. Alzheimer's disease linked Aβ42 exerts product feedback inhibition on γ-secretase impairing downstream cell signaling. eLife 2024; 12:RP90690. [PMID: 39027984 PMCID: PMC11259434 DOI: 10.7554/elife.90690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024] Open
Abstract
Amyloid β (Aβ) peptides accumulating in the brain are proposed to trigger Alzheimer's disease (AD). However, molecular cascades underlying their toxicity are poorly defined. Here, we explored a novel hypothesis for Aβ42 toxicity that arises from its proven affinity for γ-secretases. We hypothesized that the reported increases in Aβ42, particularly in the endolysosomal compartment, promote the establishment of a product feedback inhibitory mechanism on γ-secretases, and thereby impair downstream signaling events. We conducted kinetic analyses of γ-secretase activity in cell-free systems in the presence of Aβ, as well as cell-based and ex vivo assays in neuronal cell lines, neurons, and brain synaptosomes to assess the impact of Aβ on γ-secretases. We show that human Aβ42 peptides, but neither murine Aβ42 nor human Aβ17-42 (p3), inhibit γ-secretases and trigger accumulation of unprocessed substrates in neurons, including C-terminal fragments (CTFs) of APP, p75, and pan-cadherin. Moreover, Aβ42 treatment dysregulated cellular homeostasis, as shown by the induction of p75-dependent neuronal death in two distinct cellular systems. Our findings raise the possibility that pathological elevations in Aβ42 contribute to cellular toxicity via the γ-secretase inhibition, and provide a novel conceptual framework to address Aβ toxicity in the context of γ-secretase-dependent homeostatic signaling.
Collapse
Affiliation(s)
| | - Utpal Das
- Department of Neurosciences, University of California San DiegoLa JollaUnited States
| | - Sam Lismont
- VIB-KU Leuven Center for Brain & Disease ResearchLeuvenBelgium
| | - Thomas Enzlein
- VIB-KU Leuven Center for Brain & Disease ResearchLeuvenBelgium
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied SciencesMannheimGermany
| | - Masato Maesako
- Department of Neurology, Massachusetts General Hospital/Harvard Medical SchoolCharlestownUnited States
| | - Mei CQ Houser
- Department of Neurology, Massachusetts General Hospital/Harvard Medical SchoolCharlestownUnited States
| | - Maria Luisa Franco
- Molecular Basis of Neurodegeneration Unit, Instituto de Biomedicina de ValenciaValenciaSpain
| | - Burcu Özcan
- VIB-KU Leuven Center for Brain & Disease ResearchLeuvenBelgium
| | | | - Dmitry Karachentsev
- Department of Neurosciences, University of California San DiegoLa JollaUnited States
| | - Ann Becker
- Department of Neurosciences, University of California San DiegoLa JollaUnited States
| | - Carsten Hopf
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied SciencesMannheimGermany
- Medical Faculty, Heidelberg UniversityHeidelbergGermany
- Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg UniversityHeidelbergGermany
| | - Marçal Vilar
- Molecular Basis of Neurodegeneration Unit, Instituto de Biomedicina de ValenciaValenciaSpain
| | - Oksana Berezovska
- Department of Neurology, Massachusetts General Hospital/Harvard Medical SchoolCharlestownUnited States
| | - William Mobley
- Department of Neurosciences, University of California San DiegoLa JollaUnited States
| | | |
Collapse
|
13
|
Caputo M, Ivanova D, Chasserot-Golaz S, Doussau F, Haeberlé AM, Royer C, Ozkan S, Ecard J, Vitale N, Cousin MA, Tóth P, Gasman S, Ory S. Phospholipid Scramblase 1 Controls Efficient Neurotransmission and Synaptic Vesicle Retrieval at Cerebellar Synapses. J Neurosci 2024; 44:e0042242024. [PMID: 38839301 PMCID: PMC11223464 DOI: 10.1523/jneurosci.0042-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/23/2024] [Accepted: 04/28/2024] [Indexed: 06/07/2024] Open
Abstract
Phospholipids (PLs) are asymmetrically distributed at the plasma membrane. This asymmetric lipid distribution is transiently altered during calcium-regulated exocytosis, but the impact of this transient remodeling on presynaptic function is currently unknown. As phospholipid scramblase 1 (PLSCR1) randomizes PL distribution between the two leaflets of the plasma membrane in response to calcium activation, we set out to determine its role in neurotransmission. We report here that PLSCR1 is expressed in cerebellar granule cells (GrCs) and that PLSCR1-dependent phosphatidylserine egress occurred at synapses in response to neuron stimulation. Synaptic transmission is impaired at GrC Plscr1 -/- synapses, and both PS egress and synaptic vesicle (SV) endocytosis are inhibited in Plscr1 -/- cultured neurons from male and female mice, demonstrating that PLSCR1 controls PL asymmetry remodeling and SV retrieval following neurotransmitter release. Altogether, our data reveal a novel key role for PLSCR1 in SV recycling and provide the first evidence that PL scrambling at the plasma membrane is a prerequisite for optimal presynaptic performance.
Collapse
Affiliation(s)
- Margherita Caputo
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg F-67000, France
| | - Daniela Ivanova
- Centre for Discovery Brain Sciences, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
- Muir Maxwell Epilepsy Centre, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| | - Sylvette Chasserot-Golaz
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg F-67000, France
| | - Frédéric Doussau
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg F-67000, France
| | - Anne-Marie Haeberlé
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg F-67000, France
| | - Cathy Royer
- Plateforme Imagerie In Vitro, Centre National de la Recherche Scientifique UPS3256, Strasbourg F-67000, France
| | - Sebahat Ozkan
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg F-67000, France
| | - Jason Ecard
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg F-67000, France
| | - Nicolas Vitale
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg F-67000, France
| | - Michael A Cousin
- Centre for Discovery Brain Sciences, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
- Muir Maxwell Epilepsy Centre, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
- Simons Initiative for the Developing Brain, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| | - Petra Tóth
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg F-67000, France
| | - Stéphane Gasman
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg F-67000, France
| | - Stéphane Ory
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg F-67000, France
| |
Collapse
|
14
|
Simões de Oliveira L, O'Leary HE, Nawaz S, Loureiro R, Davenport EC, Baxter P, Louros SR, Dando O, Perkins E, Peltier J, Trost M, Osterweil EK, Hardingham GE, Cousin MA, Chattarji S, Booker SA, Benke TA, Wyllie DJA, Kind PC. Enhanced hippocampal LTP but normal NMDA receptor and AMPA receptor function in a rat model of CDKL5 deficiency disorder. Mol Autism 2024; 15:28. [PMID: 38877552 PMCID: PMC11177379 DOI: 10.1186/s13229-024-00601-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 05/07/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND Mutations in the X-linked gene cyclin-dependent kinase-like 5 (CDKL5) cause a severe neurological disorder characterised by early-onset epileptic seizures, autism and intellectual disability (ID). Impaired hippocampal function has been implicated in other models of monogenic forms of autism spectrum disorders and ID and is often linked to epilepsy and behavioural abnormalities. Many individuals with CDKL5 deficiency disorder (CDD) have null mutations and complete loss of CDKL5 protein, therefore in the current study we used a Cdkl5-/y rat model to elucidate the impact of CDKL5 loss on cellular excitability and synaptic function of CA1 pyramidal cells (PCs). We hypothesised abnormal pre and/or post synaptic function and plasticity would be observed in the hippocampus of Cdkl5-/y rats. METHODS To allow cross-species comparisons of phenotypes associated with the loss of CDKL5, we generated a loss of function mutation in exon 8 of the rat Cdkl5 gene and assessed the impact of the loss of CDLK5 using a combination of extracellular and whole-cell electrophysiological recordings, biochemistry, and histology. RESULTS Our results indicate that CA1 hippocampal long-term potentiation (LTP) is enhanced in slices prepared from juvenile, but not adult, Cdkl5-/y rats. Enhanced LTP does not result from changes in NMDA receptor function or subunit expression as these remain unaltered throughout development. Furthermore, Ca2+ permeable AMPA receptor mediated currents are unchanged in Cdkl5-/y rats. We observe reduced mEPSC frequency accompanied by increased spine density in basal dendrites of CA1 PCs, however we find no evidence supporting an increase in silent synapses when assessed using a minimal stimulation protocol in slices. Additionally, we found no change in paired-pulse ratio, consistent with normal release probability at Schaffer collateral to CA1 PC synapses. CONCLUSIONS Our data indicate a role for CDKL5 in hippocampal synaptic function and raise the possibility that altered intracellular signalling rather than synaptic deficits contribute to the altered plasticity. LIMITATIONS This study has focussed on the electrophysiological and anatomical properties of hippocampal CA1 PCs across early postnatal development. Studies involving other brain regions, older animals and behavioural phenotypes associated with the loss of CDKL5 are needed to understand the pathophysiology of CDD.
Collapse
MESH Headings
- Animals
- Male
- Rats
- CA1 Region, Hippocampal/metabolism
- CA1 Region, Hippocampal/pathology
- CA1 Region, Hippocampal/physiopathology
- Disease Models, Animal
- Epileptic Syndromes/genetics
- Epileptic Syndromes/metabolism
- Excitatory Postsynaptic Potentials
- Genetic Diseases, X-Linked/genetics
- Genetic Diseases, X-Linked/metabolism
- Genetic Diseases, X-Linked/physiopathology
- Hippocampus/metabolism
- Long-Term Potentiation
- Protein Serine-Threonine Kinases/metabolism
- Protein Serine-Threonine Kinases/genetics
- Pyramidal Cells/metabolism
- Pyramidal Cells/pathology
- Receptors, AMPA/metabolism
- Receptors, AMPA/genetics
- Receptors, N-Methyl-D-Aspartate/metabolism
- Receptors, N-Methyl-D-Aspartate/genetics
- Spasms, Infantile/genetics
- Spasms, Infantile/metabolism
- Synapses/metabolism
Collapse
Affiliation(s)
- Laura Simões de Oliveira
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK
| | - Heather E O'Leary
- School of Medicine, University of Colorado, Denver, CO, USA
- Department of Pharmacology, University of Colorado Denver, 12800 East 19th Ave, Aurora, CO, 80045, USA
| | - Sarfaraz Nawaz
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK
- National Centre for Biological Sciences, Tata Institute for Fundamental Research, Bangalore, 560065, India
- Centre for Brain Development and Repair, Instem, Bangalore, India
| | - Rita Loureiro
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK
| | | | - Paul Baxter
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Susana R Louros
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK
| | - Owen Dando
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Emma Perkins
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK
| | - Julien Peltier
- Faculty of Medical Sciences, Newcastle University Biosciences Institute, Newcastle upon Tyne, NE2 4HH, UK
| | - Matthias Trost
- Faculty of Medical Sciences, Newcastle University Biosciences Institute, Newcastle upon Tyne, NE2 4HH, UK
| | - Emily K Osterweil
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK
| | - Giles E Hardingham
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Michael A Cousin
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK
- Centre for Brain Development and Repair, Instem, Bangalore, India
| | - Sumantra Chattarji
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK
- National Centre for Biological Sciences, Tata Institute for Fundamental Research, Bangalore, 560065, India
- Centre for Brain Development and Repair, Instem, Bangalore, India
| | - Sam A Booker
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK
| | - Tim A Benke
- School of Medicine, University of Colorado, Denver, CO, USA.
- Department of Pharmacology, University of Colorado Denver, 12800 East 19th Ave, Aurora, CO, 80045, USA.
| | - David J A Wyllie
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK.
- Centre for Brain Development and Repair, Instem, Bangalore, India.
- Centre for Discovery Brain Sciences, Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh, EH8 9XD, UK.
| | - Peter C Kind
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK.
- Centre for Brain Development and Repair, Instem, Bangalore, India.
- Centre for Discovery Brain Sciences, Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh, EH8 9XD, UK.
| |
Collapse
|
15
|
Trease AJ, Totusek S, Lichter EZ, Stauch KL, Fox HS. Mitochondrial DNA Instability Supersedes Parkin Mutations in Driving Mitochondrial Proteomic Alterations and Functional Deficits in Polg Mutator Mice. Int J Mol Sci 2024; 25:6441. [PMID: 38928146 PMCID: PMC11203920 DOI: 10.3390/ijms25126441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Mitochondrial quality control is essential in mitochondrial function. To examine the importance of Parkin-dependent mechanisms in mitochondrial quality control, we assessed the impact of modulating Parkin on proteome flux and mitochondrial function in a context of reduced mtDNA fidelity. To accomplish this, we crossed either the Parkin knockout mouse or ParkinW402A knock-in mouse lines to the Polg mitochondrial mutator line to generate homozygous double mutants. In vivo longitudinal isotopic metabolic labeling was followed by isolation of liver mitochondria and synaptic terminals from the brain, which are rich in mitochondria. Mass spectrometry and bioenergetics analysis were assessed. We demonstrate that slower mitochondrial protein turnover is associated with loss of mtDNA fidelity in liver mitochondria but not synaptic terminals, and bioenergetic function in both tissues is impaired. Pathway analysis revealed loss of mtDNA fidelity is associated with disturbances of key metabolic pathways, consistent with its association with metabolic disorders and neurodegeneration. Furthermore, we find that loss of Parkin leads to exacerbation of Polg-driven proteomic consequences, though it may be bioenergetically protective in tissues exhibiting rapid mitochondrial turnover. Finally, we provide evidence that, surprisingly, dis-autoinhibition of Parkin (ParkinW402A) functionally resembles Parkin knockout and fails to rescue deleterious Polg-driven effects. Our study accomplishes three main outcomes: (1) it supports recent studies suggesting that Parkin dependence is low in response to an increased mtDNA mutational load, (2) it provides evidence of a potential protective role of Parkin insufficiency, and (3) it draws into question the therapeutic attractiveness of enhancing Parkin function.
Collapse
Affiliation(s)
- Andrew J. Trease
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.J.T.); (S.T.); (K.L.S.)
| | - Steven Totusek
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.J.T.); (S.T.); (K.L.S.)
| | - Eliezer Z. Lichter
- Computational Biomedicine Section, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA;
| | - Kelly L. Stauch
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.J.T.); (S.T.); (K.L.S.)
| | - Howard S. Fox
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.J.T.); (S.T.); (K.L.S.)
| |
Collapse
|
16
|
Ahmadi Asouri S, Asemi R, Aghadavod E, Jamilian M. The effect of coenzyme Q10 intake on metabolic profiles in women candidates for in-vitro fertilization: a randomised trial. Ann Med Surg (Lond) 2024; 86:3378-3384. [PMID: 38846853 PMCID: PMC11152844 DOI: 10.1097/ms9.0000000000001732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/08/2024] [Indexed: 06/09/2024] Open
Abstract
Objective Infertility and the pathogenesis of polycystic ovarian syndrome (PCOS) are both influenced by insulin resistance and dyslipidemia. Presumably, adding coenzyme Q10 (CoQ10) to these patients' diets will be beneficial. Therefore, this study aimed to examine the effects of CoQ10 supplementation on metabolic profiles in women candidates for in-vitro fertilization (IVF). Trial design and methods For this randomized, double-blinded, parallel, placebo-controlled clinical experiment, 40 PCOS-positive infertile women who were IVF candidates were included. They ranged in age from 18 to 40. The 20 participants in the two intervention groups received either CoQ10 or a placebo for 8 weeks. The expression of glucose transporter 1 (GLUT-1), peroxisome proliferator-activated receptor gamma (PPAR-γ), low-density lipoprotein receptor (LDLR), as well as metabolic profiles such as insulin metabolism and lipid profiles were evaluated. Quantitative RT-PCR determined the expression of GLUT-1, PPAR-γ, and LDLR on peripheral blood mononuclear cells. Lipid profiles and fasting glucose were assessed using enzymatic kits, and insulin was determined using Elisa kit. Results In comparison to the placebo, CoQ10 supplementation significantly reduced blood insulin levels (-0.3±1.0 vs. 0.5±0.7, P=0.01) and insulin resistance (-0.1±0.2 vs. 0.1±0.2, P=0.01), and increased PPAR-γ expression (P=0.01). In infertile PCOS patients' candidates for IVF, CoQ10 supplementation showed no appreciable impact on other metabolic profiles. Also, CoQ10 supplementation revealed no significant impact on GLUT-1 (P=0.30), or LDLR (P=0.27) expression. Within-group changes in insulin levels (P=0.01) and insulin resistance (P=0.01) showed a significant elevation in the placebo group. When we adjusted the analysis for baseline BMI, baseline values of variables, and age, our findings were not affected. Conclusions Eight weeks of CoQ10 supplementation demonstrated positive benefits on PPAR-γ expression, insulin resistance, and serum insulin in infertile PCOS women candidates for IVF.
Collapse
Affiliation(s)
- Sahar Ahmadi Asouri
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan
| | - Reza Asemi
- Department of Internal Medicine, School of Medicine, Cancer Prevention Research Center, Seyyed Al-Shohada Hospital, Isfahan University of Medical Sciences, Isfahan
| | - Esmat Aghadavod
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan
| | - Mehri Jamilian
- Traditional and Complementary Medicine Research Center, Arak University of Medical Sciences, Arak, Iran
| |
Collapse
|
17
|
Lu CW, Lin TY, Chiu KM, Lee MY, Wang SJ. Gypenoside XVII Reduces Synaptic Glutamate Release and Protects against Excitotoxic Injury in Rats. Biomolecules 2024; 14:589. [PMID: 38785996 PMCID: PMC11118014 DOI: 10.3390/biom14050589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/15/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
Excitotoxicity is a common pathological process in neurological diseases caused by excess glutamate. The purpose of this study was to evaluate the effect of gypenoside XVII (GP-17), a gypenoside monomer, on the glutamatergic system. In vitro, in rat cortical nerve terminals (synaptosomes), GP-17 dose-dependently decreased glutamate release with an IC50 value of 16 μM. The removal of extracellular Ca2+ or blockade of N-and P/Q-type Ca2+ channels and protein kinase A (PKA) abolished the inhibitory effect of GP-17 on glutamate release from cortical synaptosomes. GP-17 also significantly reduced the phosphorylation of PKA, SNAP-25, and synapsin I in cortical synaptosomes. In an in vivo rat model of glutamate excitotoxicity induced by kainic acid (KA), GP-17 pretreatment significantly prevented seizures and rescued neuronal cell injury and glutamate elevation in the cortex. GP-17 pretreatment decreased the expression levels of sodium-coupled neutral amino acid transporter 1, glutamate synthesis enzyme glutaminase and vesicular glutamate transporter 1 but increased the expression level of glutamate metabolism enzyme glutamate dehydrogenase in the cortex of KA-treated rats. In addition, the KA-induced alterations in the N-methyl-D-aspartate receptor subunits GluN2A and GluN2B in the cortex were prevented by GP-17 pretreatment. GP-17 also prevented the KA-induced decrease in cerebral blood flow and arginase II expression. These results suggest that (i) GP-17, through the suppression of N- and P/Q-type Ca2+ channels and consequent PKA-mediated SNAP-25 and synapsin I phosphorylation, reduces glutamate exocytosis from cortical synaptosomes; and (ii) GP-17 has a neuroprotective effect on KA-induced glutamate excitotoxicity in rats through regulating synaptic glutamate release and cerebral blood flow.
Collapse
Affiliation(s)
- Cheng-Wei Lu
- Department of Anesthesiology, Far-Eastern Memorial Hospital, New Taipei 22060, Taiwan; (C.-W.L.); (T.-Y.L.)
- Department of Mechanical Engineering, Yuan Ze University, Taoyuan 32003, Taiwan
| | - Tzu-Yu Lin
- Department of Anesthesiology, Far-Eastern Memorial Hospital, New Taipei 22060, Taiwan; (C.-W.L.); (T.-Y.L.)
- Department of Mechanical Engineering, Yuan Ze University, Taoyuan 32003, Taiwan
| | - Kuan-Ming Chiu
- Division of Cardiovascular Surgery, Cardiovascular Center, Far-Eastern Memorial Hospital, New Taipei 22060, Taiwan;
- Department of Electrical Engineering, Yuan Ze University, Taoyuan 32003, Taiwan
| | - Ming-Yi Lee
- Department of Medical Research, Far-Eastern Memorial Hospital, New Taipei 22060, Taiwan;
| | - Su-Jane Wang
- School of Medicine, Fu Jen Catholic University, New Taipei 24205, Taiwan
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan
| |
Collapse
|
18
|
Li D, Gao X, Ma X, Wang M, Cheng C, Xue T, Gao F, Shen Y, Zhang J, Liu Q. Aging-induced tRNA Glu-derived fragment impairs glutamate biosynthesis by targeting mitochondrial translation-dependent cristae organization. Cell Metab 2024; 36:1059-1075.e9. [PMID: 38458203 DOI: 10.1016/j.cmet.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 10/31/2023] [Accepted: 02/15/2024] [Indexed: 03/10/2024]
Abstract
Mitochondrial cristae, infoldings of the mitochondrial inner membrane, undergo aberrant changes in their architecture with age. However, the underlying molecular mechanisms and their contribution to brain aging are largely elusive. Here, we observe an age-dependent accumulation of Glu-5'tsRNA-CTC, a transfer-RNA-derived small RNA (tsRNA), derived from nuclear-encoded tRNAGlu in the mitochondria of glutaminergic neurons. Mitochondrial Glu-5'tsRNA-CTC disrupts the binding of mt-tRNALeu and leucyl-tRNA synthetase2 (LaRs2), impairing mt-tRNALeu aminoacylation and mitochondria-encoded protein translation. Mitochondrial translation defects disrupt cristae organization, leading to damaged glutaminase (GLS)-dependent glutamate formation and reduced synaptosomal glutamate levels. Moreover, reduction of Glu-5'tsRNA-CTC protects aged brains from age-related defects in mitochondrial cristae organization, glutamate metabolism, synaptic structures, and memory. Thus, beyond illustrating a physiological role for normal mitochondrial cristae ultrastructure in maintaining glutamate levels, our study defines a pathological role for tsRNAs in brain aging and age-related memory decline.
Collapse
Affiliation(s)
- Dingfeng Li
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China; Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Xinyi Gao
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China; Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Xiaolin Ma
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China; Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Ming Wang
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Chuandong Cheng
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Tian Xue
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Feng Gao
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China
| | - Yong Shen
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China; Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Neurodegenerative Disorder Research Center, Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei 230026, China
| | - Juan Zhang
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China; Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Qiang Liu
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China; Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Neurodegenerative Disorder Research Center, Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei 230026, China; Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei 230001, China.
| |
Collapse
|
19
|
Zoltowska KM, Das U, Lismont S, Enzlein T, Maesako M, Houser MCQ, Franco ML, Özcan B, Moreira DG, Karachentsev D, Becker A, Hopf C, Vilar M, Berezovska O, Mobley W, Chávez-Gutiérrez L. Alzheimer's disease linked Aβ42 exerts product feedback inhibition on γ-secretase impairing downstream cell signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.02.551596. [PMID: 37577527 PMCID: PMC10418207 DOI: 10.1101/2023.08.02.551596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Amyloid β (Aβ) peptides accumulating in the brain are proposed to trigger Alzheimer's disease (AD). However, molecular cascades underlying their toxicity are poorly defined. Here, we explored a novel hypothesis for Aβ42 toxicity that arises from its proven affinity for γ-secretases. We hypothesized that the reported increases in Aβ42, particularly in the endolysosomal compartment, promote the establishment of a product feedback inhibitory mechanism on γ-secretases, and thereby impair downstream signaling events. We show that human Aβ42 peptides, but neither murine Aβ42 nor human Aβ17-42 (p3), inhibit γ-secretases and trigger accumulation of unprocessed substrates in neurons, including C-terminal fragments (CTFs) of APP, p75 and pan-cadherin. Moreover, Aβ42 treatment dysregulated cellular -homeostasis, as shown by the induction of p75-dependent neuronal death in two distinct cellular systems. Our findings raise the possibility that pathological elevations in Aβ42 contribute to cellular toxicity via the γ-secretase inhibition, and provide a novel conceptual framework to address Aβ toxicity in the context of γ-secretase-dependent homeostatic signaling.
Collapse
Affiliation(s)
| | - Utpal Das
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States of America
| | - Sam Lismont
- VIB-KU Leuven Center for Brain & Disease Research, VIB, Leuven, Belgium
| | - Thomas Enzlein
- VIB-KU Leuven Center for Brain & Disease Research, VIB, Leuven, Belgium
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Mannheim, Germany
| | - Masato Maesako
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown, MA, United States of America
| | - Mei CQ Houser
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown, MA, United States of America
| | - María Luisa Franco
- Molecular Basis of Neurodegeneration Unit, Institute of Biomedicine of València (IBV-CSIC), València, Spain
| | - Burcu Özcan
- VIB-KU Leuven Center for Brain & Disease Research, VIB, Leuven, Belgium
| | | | - Dmitry Karachentsev
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States of America
| | - Ann Becker
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States of America
| | - Carsten Hopf
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Mannheim, Germany
- Medical Faculty, Heidelberg University, Heidelberg, Germany
- Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Marçal Vilar
- Molecular Basis of Neurodegeneration Unit, Institute of Biomedicine of València (IBV-CSIC), València, Spain
| | - Oksana Berezovska
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown, MA, United States of America
| | - William Mobley
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States of America
| | | |
Collapse
|
20
|
Hill LJ, Messias CSMDA, Vilela CLS, Garritano AN, Villela HDM, do Carmo FL, Thomas T, Peixoto RS. Bacteria associated with the in hospite Symbiodiniaceae's phycosphere. iScience 2024; 27:109531. [PMID: 38585661 PMCID: PMC10995889 DOI: 10.1016/j.isci.2024.109531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 12/23/2023] [Accepted: 03/16/2024] [Indexed: 04/09/2024] Open
Abstract
Symbiotic interactions between Symbiodiniaceae and bacteria are still poorly explored, especially those in hospite. Here, we adapted a technique that allows for the enrichment of intact and metabolically active in hospite Symbiodiniaceae cells (ihSC) and their associated bacteria from the tissue of the model coral Pocillopora damicornis, using a discontinuous gradient of solution of isotonic Percoll (SIP). The ihSC were concentrated in the 50% SIP fraction, as determined by microscopy. The presence of bacteria associated with ihSC was confirmed by fluorescence in situ hybridization, while microbiome analysis indicated that bacteria of the families Halieaceae, Flavobacteriaceae, and Alcanivoraceae are significantly associated with ihSC. Extracellular vesicles that could be exuding molecules were detected on the symbiosome membranes. Our technique and data contribute to elucidate ihSC-bacteria interactions.
Collapse
Affiliation(s)
- Lilian Jorge Hill
- Laboratory of Molecular Microbial Ecology, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | | | - Caren Leite Spindola Vilela
- Laboratory of Molecular Microbial Ecology, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Alessandro N Garritano
- Centre for Marine Science and Innovation, School of Biological, Earth and Environmental Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Helena Dias Muller Villela
- Red Sea Research Center, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal 23955, Kingdom of Saudi Arabia
| | - Flavia Lima do Carmo
- Laboratory of Molecular Microbial Ecology, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Torsten Thomas
- Centre for Marine Science and Innovation, School of Biological, Earth and Environmental Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Raquel S. Peixoto
- Red Sea Research Center, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal 23955, Kingdom of Saudi Arabia
| |
Collapse
|
21
|
Sun C. Single-Molecule-Resolution Approaches in Synaptic Biology. J Phys Chem B 2024; 128:3061-3068. [PMID: 38513216 DOI: 10.1021/acs.jpcb.3c08026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
Synapses between neurons are the primary loci for information transfer and storage in the brain. An individual neuron, alone, can make over 10000 synaptic contacts. It is, however, not easy to investigate what goes on locally within a synapse because many synaptic compartments are only a few hundred nanometers wide in size─close to the diffraction limit of light. To observe the biomolecular machinery and processes within synapses, in situ single-molecule techniques are emerging as powerful tools. Guided by important biological questions, this Perspective will highlight recent advances in using these techniques to obtain in situ measurements of synaptic molecules in three aspects: the cell-biological machinery within synapses, the synaptic architecture, and the synaptic neurotransmitter receptors. These advances showcase the increasing importance of single-molecule-resolution techniques for accessing subcellular biophysical and biomolecular information related to the brain.
Collapse
Affiliation(s)
- Chao Sun
- Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen 81, 8000 Aarhus C, Denmark
| |
Collapse
|
22
|
Olivero G, Taddeucci A, Vallarino G, Trebesova H, Roggeri A, Gagliani MC, Cortese K, Grilli M, Pittaluga A. Complement tunes glutamate release and supports synaptic impairments in an animal model of multiple sclerosis. Br J Pharmacol 2024. [PMID: 38369641 DOI: 10.1111/bph.16328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 12/05/2023] [Accepted: 01/03/2024] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND AND PURPOSE To deepen our knowledge of the role of complement in synaptic impairment in experimental autoimmune encephalomyelitis (EAE) mice, we investigated the distribution of C1q and C3 proteins and the role of complement as a promoter of glutamate release in purified nerve endings (synaptosomes) and astrocytic processes (gliosomes) isolated from the cortex of EAE mice at the acute stage of the disease (21 ± 1 day post-immunization). EXPERIMENTAL APPROACH EAE cortical synaptosomes and gliosomes were analysed for glutamate release efficiency (measured as release of preloaded [3 H]D-aspartate ([3 H]D-ASP)), C1q and C3 protein density, and for viability and ongoing apoptosis. KEY RESULTS In healthy mice, complement releases [3 H]D-ASP from gliosomes more efficiently than from synaptosomes. The releasing activity occurs in a dilution-dependent manner and involves the reversal of the excitatory amino acid transporters (EAATs). In EAE mice, the complement-induced releasing activity is significantly reduced in cortical synaptosomes but amplified in cortical gliosomes. These adaptations are paralleled by decreased density of the EAAT2 protein in synaptosomes and increased EAAT1 staining in gliosomes. Concomitantly, PSD95, GFAP, and CD11b, but not SNAP25, proteins are overexpressed in the cortex of the EAE mice. Similarly, C1q and C3 protein immunostaining is increased in EAE cortical synaptosomes and gliosomes, although signs of ongoing apoptosis or altered viability are not detectable. CONCLUSION AND IMPLICATIONS Our results unveil a new noncanonical role of complement in the CNS of EAE mice relevant to disease progression and central synaptopathy that suggests new therapeutic targets for the management of MS.
Collapse
Affiliation(s)
- Guendalina Olivero
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | - Alice Taddeucci
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | - Giulia Vallarino
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | - Hanna Trebesova
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | - Alessandra Roggeri
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | - Maria Cristina Gagliani
- DIMES, Department of Experimental Medicine, Cellular Electron Microscopy Laboratory, Università di Genova, Genoa, Italy
| | - Katia Cortese
- DIMES, Department of Experimental Medicine, Cellular Electron Microscopy Laboratory, Università di Genova, Genoa, Italy
| | - Massimo Grilli
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | - Anna Pittaluga
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, Centre of Excellence for Biomedical Research, 3Rs Center, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
23
|
Wegener AJ, Hyer MM, Targett I, Kloster A, Shaw GA, Rodriguez AMM, Dyer SK, Neigh GN. Behavior, synaptic mitochondria, and microglia are differentially impacted by chronic adolescent stress and repeated endotoxin exposure in male and female rats. Stress 2024; 27:2299971. [PMID: 38179979 PMCID: PMC11064104 DOI: 10.1080/10253890.2023.2299971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 12/16/2023] [Indexed: 01/06/2024] Open
Abstract
Early life adversity and chronic inflammation have both been associated with cognitive impairment and neural compromise. In this study, we investigated the interactions between a history of chronic adolescent stress (CAS) and repeated endotoxin exposure on behavior, synaptic mitochondria, and microglia in adult male and female Wistar rats. Adult rats from chronic stress and control conditions were exposed to either repeated endotoxin (lipopolysaccharide; LPS) or saline injections every 3 days for 9 weeks. In both sexes, repeated LPS, regardless of stress history, impaired working memory in the Y maze. Regarding spatial memory, LPS impaired function for females; whereas, CAS altered function in males. Although males had an increase in anxiety-like behavior shortly after CAS, there were no long-term effects on anxiety-like behavior or social interaction observed in males or females. Stress did not alter synaptic mitochondrial function in either sex. Repeated LPS altered synaptic mitochondrial function such that ATP production was increased in females only. There were no observed increases in IBA-1 positive cells within the hippocampus for either sex. However, LPS and CAS altered microglia morphology in females. Impact of repeated LPS was evident at the terminal endpoint with increased spleen weight in both sexes and decreased adrenal weight in males only. Circulating cytokines were not impacted by repeated LPS at the terminal endpoint, but evidence of CAS effects on cytokines in females were evident. These data suggest a long-term impact of chronic stress and an impact of repeated endotoxin challenge in adulthood; however, not all physiological and behavioral metrics examined were impacted by the paradigm employed in this study and the two environmental challenges rarely interacted.
Collapse
Affiliation(s)
- A J Wegener
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - M M Hyer
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - I Targett
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - A Kloster
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - G A Shaw
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - A M M Rodriguez
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - S K Dyer
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - G N Neigh
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
24
|
Martínez-Aguirre C, Márquez LA, Santiago-Castañeda CL, Carmona-Cruz F, Nuñez-Lumbreras MDLA, Martínez-Rojas VA, Alonso-Vanegas M, Aguado-Carrillo G, Gómez-Víquez NL, Galván EJ, Cuéllar-Herrera M, Rocha L. Cannabidiol Modifies the Glutamate Over-Release in Brain Tissue of Patients and Rats with Epilepsy: A Pilot Study. Biomedicines 2023; 11:3237. [PMID: 38137458 PMCID: PMC10741033 DOI: 10.3390/biomedicines11123237] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
Drug-resistant epilepsy (DRE) is associated with high extracellular levels of glutamate. Studies support the idea that cannabidiol (CBD) decreases glutamate over-release. This study focused on investigating whether CBD reduces the evoked glutamate release in cortical synaptic terminals obtained from patients with DRE as well as in a preclinical model of epilepsy. Synaptic terminals (synaptosomes) were obtained from the epileptic neocortex of patients with drug-resistant temporal lobe epilepsy (DR-TLE, n = 10) or drug-resistant extratemporal lobe epilepsy (DR-ETLE, n = 10) submitted to epilepsy surgery. Synaptosomes highly purified by Percoll-sucrose density gradient were characterized by confocal microscopy and Western blot. Synaptosomes were used to estimate the high KCl (33 mM)-evoked glutamate release in the presence of CBD at different concentrations. Our results revealed responsive tissue obtained from seven patients with DR-TLE and seven patients with DR-ETLE. Responsive tissue showed lower glutamate release (p < 0.05) when incubated with CBD at low concentrations (less than 100 µM) but not at higher concentrations. Tissue that was non-responsive to CBD (DR-TLE, n = 3 and DR-ELTE, n = 3) showed high glutamate release despite CBD exposure at different concentrations. Simultaneously, a block of the human epileptic neocortex was used to determine its viability through whole-cell and extracellular electrophysiological recordings. The electrophysiological evaluations supported that the responsive and non-responsive human epileptic neocortices used in the present study exhibited proper neuronal viability and stability to acquire electrophysiological responses. We also investigated whether the subchronic administration of CBD could reduce glutamate over-release in a preclinical model of temporal lobe epilepsy. Administration of CBD (200 mg/kg, p.o. every 24 h for 7 days) to rats with lithium-pilocarpine-evoked spontaneous recurrent seizures reduced glutamate over-release in the hippocampus. The present study revealed that acute exposure to low concentrations of CBD can reduce the glutamate over-release in synaptic terminals obtained from some patients with DRE. This effect is also evident when applied subchronically in rats with spontaneous recurrent seizures. An important finding was the identification of a group of patients that were non-responsive to CBD effects. Future studies are essential to identify biomarkers of responsiveness to CBD to control DRE.
Collapse
Affiliation(s)
- Christopher Martínez-Aguirre
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
| | - Luis Alfredo Márquez
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
| | - Cindy Lizbeth Santiago-Castañeda
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
| | - Francia Carmona-Cruz
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
| | - Maria de los Angeles Nuñez-Lumbreras
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
| | - Vladimir A. Martínez-Rojas
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
- Center for Research on Aging, Center for Research and Advanced Studies, Mexico City 14330, Mexico
| | - Mario Alonso-Vanegas
- International Center for Epilepsy Surgery, HMG-Coyoacán Hospital, Mexico City 04380, Mexico;
| | - Gustavo Aguado-Carrillo
- Clinic of Epilepsy, General Hospital of México Dr. Eduardo Liceaga, Mexico City 06720, Mexico
| | - Norma L. Gómez-Víquez
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
| | - Emilio J. Galván
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
- Center for Research on Aging, Center for Research and Advanced Studies, Mexico City 14330, Mexico
| | - Manola Cuéllar-Herrera
- Clinic of Epilepsy, General Hospital of México Dr. Eduardo Liceaga, Mexico City 06720, Mexico
| | - Luisa Rocha
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
| |
Collapse
|
25
|
Bowen ER, DiGiacomo P, Fraser HP, Guttenplan K, Smith BAH, Heberling ML, Vidano L, Shah N, Shamloo M, Wilson JL, Grimes KV. Beta-2 adrenergic receptor agonism alters astrocyte phagocytic activity and has potential applications to psychiatric disease. DISCOVER MENTAL HEALTH 2023; 3:27. [PMID: 38036718 PMCID: PMC10689618 DOI: 10.1007/s44192-023-00050-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 10/26/2023] [Indexed: 12/02/2023]
Abstract
Schizophrenia is a debilitating condition necessitating more efficacious therapies. Previous studies suggested that schizophrenia development is associated with aberrant synaptic pruning by glial cells. We pursued an interdisciplinary approach to understand whether therapeutic reduction in glial cell-specifically astrocytic-phagocytosis might benefit neuropsychiatric patients. We discovered that beta-2 adrenergic receptor (ADRB2) agonists reduced phagocytosis using a high-throughput, phenotypic screen of over 3200 compounds in primary human fetal astrocytes. We used protein interaction pathways analysis to associate ADRB2, to schizophrenia and endocytosis. We demonstrated that patients with a pediatric exposure to salmeterol, an ADRB2 agonist, had reduced in-patient psychiatry visits using a novel observational study in the electronic health record. We used a mouse model of inflammatory neurodegenerative disease and measured changes in proteins associated with endocytosis and vesicle-mediated transport after ADRB2 agonism. These results provide substantial rationale for clinical consideration of ADRB2 agonists as possible therapies for patients with schizophrenia.
Collapse
Affiliation(s)
- Ellen R Bowen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Weill Cornell Medicine, New York, NY, USA
- University of Michigan Medical School, Ann Arbor, MI, USA
| | - Phillip DiGiacomo
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Hannah P Fraser
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kevin Guttenplan
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Benjamin A H Smith
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Marlene L Heberling
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Laura Vidano
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Nigam Shah
- Center for Biomedical Informatics Research, Stanford School of Medicine, Stanford, CA, USA
| | - Mehrdad Shamloo
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Jennifer L Wilson
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA.
| | - Kevin V Grimes
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
26
|
Liau WS, Zhao Q, Bademosi A, Gormal RS, Gong H, Marshall PR, Periyakaruppiah A, Madugalle SU, Zajaczkowski EL, Leighton LJ, Ren H, Musgrove M, Davies J, Rauch S, He C, Dickinson BC, Li X, Wei W, Meunier FA, Fernández-Moya SM, Kiebler MA, Srinivasan B, Banerjee S, Clark M, Spitale RC, Bredy TW. Fear extinction is regulated by the activity of long noncoding RNAs at the synapse. Nat Commun 2023; 14:7616. [PMID: 37993455 PMCID: PMC10665438 DOI: 10.1038/s41467-023-43535-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 11/12/2023] [Indexed: 11/24/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) represent a multidimensional class of regulatory molecules that are involved in many aspects of brain function. Emerging evidence indicates that lncRNAs are localized to the synapse; however, a direct role for their activity in this subcellular compartment in memory formation has yet to be demonstrated. Using lncRNA capture-seq, we identified a specific set of lncRNAs that accumulate in the synaptic compartment within the infralimbic prefrontal cortex of adult male C57/Bl6 mice. Among these was a splice variant related to the stress-associated lncRNA, Gas5. RNA immunoprecipitation followed by mass spectrometry and single-molecule imaging revealed that this Gas5 isoform, in association with the RNA binding proteins G3BP2 and CAPRIN1, regulates the activity-dependent trafficking and clustering of RNA granules. In addition, we found that cell-type-specific, activity-dependent, and synapse-specific knockdown of the Gas5 variant led to impaired fear extinction memory. These findings identify a new mechanism of fear extinction that involves the dynamic interaction between local lncRNA activity and RNA condensates in the synaptic compartment.
Collapse
Affiliation(s)
- Wei-Siang Liau
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia.
| | - Qiongyi Zhao
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Adekunle Bademosi
- Single Molecule Neuroscience Laboratory, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Rachel S Gormal
- Single Molecule Neuroscience Laboratory, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Hao Gong
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Paul R Marshall
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Ambika Periyakaruppiah
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Sachithrani U Madugalle
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Esmi L Zajaczkowski
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Laura J Leighton
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Haobin Ren
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Mason Musgrove
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Joshua Davies
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Simone Rauch
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
| | - Chuan He
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
| | - Bryan C Dickinson
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
| | - Xiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Medical Research Institute, Wuhan University, Wuhan, China
| | - Wei Wei
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Frédéric A Meunier
- Single Molecule Neuroscience Laboratory, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Sandra M Fernández-Moya
- Biomedical Centre, Ludwig Maximilian University of Munich, Munich, Germany
- Gene Regulation of Cell Identity, Regenerative Medicine Program, Bellvitge Institute for Biomedical Research (IDIBELL) and Program for Advancing Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L'Hospitalet del Llobregat, 08908, Barcelona, Spain
| | - Michael A Kiebler
- Biomedical Centre, Ludwig Maximilian University of Munich, Munich, Germany
| | | | | | - Michael Clark
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Robert C Spitale
- Department of Pharmaceutical Sciences, The University of California, Irvine, CA, USA
| | - Timothy W Bredy
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
27
|
van Oostrum M, Blok TM, Giandomenico SL, Tom Dieck S, Tushev G, Fürst N, Langer JD, Schuman EM. The proteomic landscape of synaptic diversity across brain regions and cell types. Cell 2023; 186:5411-5427.e23. [PMID: 37918396 PMCID: PMC10686415 DOI: 10.1016/j.cell.2023.09.028] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 08/18/2023] [Accepted: 09/28/2023] [Indexed: 11/04/2023]
Abstract
Neurons build synaptic contacts using different protein combinations that define the specificity, function, and plasticity potential of synapses; however, the diversity of synaptic proteomes remains largely unexplored. We prepared synaptosomes from 7 different transgenic mouse lines with fluorescently labeled presynaptic terminals. Combining microdissection of 5 different brain regions with fluorescent-activated synaptosome sorting (FASS), we isolated and analyzed the proteomes of 18 different synapse types. We discovered ∼1,800 unique synapse-type-enriched proteins and allocated thousands of proteins to different types of synapses (https://syndive.org/). We identify shared synaptic protein modules and highlight the proteomic hotspots for synapse specialization. We reveal unique and common features of the striatal dopaminergic proteome and discover the proteome signatures that relate to the functional properties of different interneuron classes. This study provides a molecular systems-biology analysis of synapses and a framework to integrate proteomic information for synapse subtypes of interest with cellular or circuit-level experiments.
Collapse
Affiliation(s)
- Marc van Oostrum
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | - Thomas M Blok
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | | | | | - Georgi Tushev
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | - Nicole Fürst
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | - Julian D Langer
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany; Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Erin M Schuman
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany.
| |
Collapse
|
28
|
Reid KM, Brown GC. LRPAP1 is released from activated microglia and inhibits microglial phagocytosis and amyloid beta aggregation. Front Immunol 2023; 14:1286474. [PMID: 38035103 PMCID: PMC10687467 DOI: 10.3389/fimmu.2023.1286474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Low-density lipoprotein receptor-related protein-associated protein 1 (LRPAP1), also known as receptor associated protein (RAP), is an endoplasmic reticulum (ER) chaperone and inhibitor of LDL receptor related protein 1 (LRP1) and related receptors. These receptors have dozens of physiological ligands and cell functions, but it is not known whether cells release LRPAP1 physiologically at levels that regulate these receptors and cell functions. We used mouse BV-2 and human CHME3 microglial cell lines, and found that microglia released nanomolar levels of LRPAP1 when inflammatory activated by lipopolysaccharide or when ER stressed by tunicamycin. LRPAP1 was found on the surface of live activated and non-activated microglia, and anti-LRPAP1 antibodies induced internalization. Addition of 10 nM LRPAP1 inhibited microglial phagocytosis of isolated synapses and cells, and the uptake of Aβ. LRPAP1 also inhibited Aβ aggregation in vitro. Thus, activated and stressed microglia release LRPAP1 levels that can inhibit phagocytosis, Aβ uptake and Aβ aggregation. We conclude that LRPAP1 release may regulate microglial functions and Aβ pathology, and more generally that extracellular LRPAP1 may be a physiological and pathological regulator of a wide range of cell functions.
Collapse
Affiliation(s)
| | - Guy C. Brown
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
29
|
Itoh N, Itoh Y, Stiles L, Voskuhl R. Sex differences in the neuronal transcriptome and synaptic mitochondrial function in the cerebral cortex of a multiple sclerosis model. Front Neurol 2023; 14:1268411. [PMID: 38020654 PMCID: PMC10654219 DOI: 10.3389/fneur.2023.1268411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Multiple sclerosis (MS) affects the cerebral cortex, inducing cortical atrophy and neuronal and synaptic pathology. Despite the fact that women are more susceptible to getting MS, men with MS have worse disability progression. Here, sex differences in neurodegenerative mechanisms are determined in the cerebral cortex using the MS model, chronic experimental autoimmune encephalomyelitis (EAE). Methods Neurons from cerebral cortex tissues of chronic EAE, as well as age-matched healthy control, male and female mice underwent RNA sequencing and gene expression analyses using RiboTag technology. The morphology of mitochondria in neurons of cerebral cortex was assessed using Thy1-CFP-MitoS mice. Oxygen consumption rates were determined using mitochondrial respirometry assays from intact as well as permeabilized synaptosomes. Results RNA sequencing of neurons in cerebral cortex during chronic EAE in C57BL/6 mice showed robust differential gene expression in male EAE compared to male healthy controls. In contrast, there were few differences in female EAE compared to female healthy controls. The most enriched differential gene expression pathways in male mice during EAE were mitochondrial dysfunction and oxidative phosphorylation. Mitochondrial morphology in neurons showed significant abnormalities in the cerebral cortex of EAE males, but not EAE females. Regarding function, synaptosomes isolated from cerebral cortex of male, but not female, EAE mice demonstrated significantly decreased oxygen consumption rates during respirometry assays. Discussion Cortical neuronal transcriptomics, mitochondrial morphology, and functional respirometry assays in synaptosomes revealed worse neurodegeneration in male EAE mice. This is consistent with worse neurodegeneration in MS men and reveals a model and a target to develop treatments to prevent cortical neurodegeneration and mitigate disability progression in MS men.
Collapse
Affiliation(s)
- Noriko Itoh
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Yuichiro Itoh
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Linsey Stiles
- Department of Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Rhonda Voskuhl
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
30
|
Hu X, Zhao HL, Kurban N, Qin Y, Chen X, Cui SY, Zhang YH. Reduction of BDNF Levels and Biphasic Changes in Glutamate Release in the Prefrontal Cortex Correlate with Susceptibility to Chronic Stress-Induced Anhedonia. eNeuro 2023; 10:ENEURO.0406-23.2023. [PMID: 37989582 PMCID: PMC10668226 DOI: 10.1523/eneuro.0406-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 10/17/2023] [Accepted: 10/26/2023] [Indexed: 11/23/2023] Open
Abstract
Chronic stress has been considered to induce depressive symptoms, such as anhedonia, particularly in susceptible individuals. Synaptic plasticity in the prefrontal cortex (PFC) is closely associated with susceptibility or resilience to chronic stress-induced anhedonia. However, effects of chronic stress with different durations on the neurobiological mechanisms that underlie susceptibility to anhedonia remain unclear. The present study investigated effects of chronic mild stress (CMS) for 14, 21, and 35 d on anhedonia-like behavior and glutamate synapses in the PFC. We found that brain-derived neurotrophic factor (BDNF) levels in the PFC significantly decreased only in anhedonia-susceptible rats that were exposed to CMS for 14, 21, and 35 d. Additionally, 14 d of CMS increased prefrontal glutamate release, and 35 d of CMS decreased glutamate release, in addition to reducing synaptic proteins and spine density in the PFC. Moreover, we found that anhedonia-like behavior in a subset of rats spontaneously decreased, accompanied by the restoration of BDNF levels and glutamate release, on day 21 of CMS. Ketamine treatment restored the reduction of BDNF levels and biphasic changes in glutamate release that were induced by CMS. Our findings revealed a progressive reduction of synaptic plasticity and biphasic changes in glutamate release in the PFC during CMS. Reductions of BDNF levels may be key neurobiological markers of susceptibility to stress-induced anhedonia.
Collapse
Affiliation(s)
- Xiao Hu
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing 100191, China
| | - Hui-Ling Zhao
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing 100191, China
| | - Nurhumar Kurban
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing 100191, China
| | - Yu Qin
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing 100191, China
| | - Xi Chen
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing 100191, China
| | - Su-Ying Cui
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing 100191, China
| | - Yong-He Zhang
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing 100191, China
| |
Collapse
|
31
|
Lombardi P, Karadayian AG, Guerra JI, Bustamante J, Rodríguez de Lores Arnaiz G, Lores-Arnaiz S. Mitochondrial bioenergetics and cytometric characterization of a synaptosomal preparation from mouse brain cortex. Mitochondrion 2023; 73:95-107. [PMID: 37944836 DOI: 10.1016/j.mito.2023.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 09/06/2023] [Accepted: 10/28/2023] [Indexed: 11/12/2023]
Abstract
Mitochondrial function at synapses can be assessed in isolated nerve terminals. Synaptosomes are structures obtained in vitro by detaching the nerve endings from neuronal bodies under controlled homogenization conditions. Several protocols have been described for the preparation of intact synaptosomal fractions. Herein a fast and economical method to obtain synaptosomes with optimal intrasynaptic mitochondria functionality was described. Synaptosomal fractions were obtained from mouse brain cortex by differential centrifugation followed by centrifugation in a Ficoll gradient. The characteristics of the subcellular particles obtained were analyzed by flow cytometry employing specific tools. Integrity and specificity of the obtained organelles were evaluated by calcein and SNAP-25 probes. The proportion of positive events of the synaptosomal preparation was 75 ± 2 % and 48 ± 7% for calcein and Synaptosomal-Associated Protein of 25 kDa (SNAP-25), respectively. Mitochondrial integrity was evaluated by flow cytometric analysis of cardiolipin content, which indicated that 73 ± 1% of the total events were 10 N-nonylacridine orange (NAO)-positive. Oxygen consumption, ATP production and mitochondrial membrane potential determinations showed that mitochondria inside synaptosomes remained functional after the isolation procedure. Mitochondrial and synaptosomal enrichment were determined by measuring synaptosomes/ homogenate ratio of specific markers. Functionality of synaptosomes was verified by nitric oxide detection after glutamate addition. As compared with other methods, the present protocol can be performed briefly, does not imply high economic costs, and provides an useful tool for the isolation of a synaptosomal preparation with high mitochondrial respiratory capacity and an adequate integrity and function of intraterminal mitochondria.
Collapse
Affiliation(s)
- Paulina Lombardi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Fisicoquímica, Buenos Aires, Argentina; CONICET- Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular "Profesor Alberto Boveris" (IBIMOL), Buenos Aires, Argentina
| | - Analía G Karadayian
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Fisicoquímica, Buenos Aires, Argentina; CONICET- Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular "Profesor Alberto Boveris" (IBIMOL), Buenos Aires, Argentina
| | - Juan I Guerra
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Fisicoquímica, Buenos Aires, Argentina; CONICET- Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular "Profesor Alberto Boveris" (IBIMOL), Buenos Aires, Argentina
| | | | - Georgina Rodríguez de Lores Arnaiz
- Universidad de Buenos Aires, Facultad de Medicina, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencias "Profesor Eduardo De Robertis" (IBCN), Buenos Aires, Argentina
| | - Silvia Lores-Arnaiz
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Fisicoquímica, Buenos Aires, Argentina; CONICET- Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular "Profesor Alberto Boveris" (IBIMOL), Buenos Aires, Argentina.
| |
Collapse
|
32
|
Madeira D, Domingues J, Lopes CR, Canas PM, Cunha RA, Agostinho P. Modification of astrocytic Cx43 hemichannel activity in animal models of AD: modulation by adenosine A 2A receptors. Cell Mol Life Sci 2023; 80:340. [PMID: 37898985 PMCID: PMC10613596 DOI: 10.1007/s00018-023-04983-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/07/2023] [Accepted: 09/26/2023] [Indexed: 10/31/2023]
Abstract
Increasing evidence implicates astrocytic dysfunction in Alzheimer's disease (AD), a neurodegenerative disorder characterised by progressive cognitive loss. The accumulation of amyloid-β (Aβ) plaques is a histopathological hallmark of AD and associated with increased astrocyte reactivity. In APP/PS1 mice modelling established AD (9 months), we now show an altered astrocytic morphology and enhanced activity of astrocytic hemichannels, mainly composed by connexin 43 (Cx43). Hemichannel activity in hippocampal astrocytes is also increased in two models of early AD: (1) mice with intracerebroventricular (icv) administration of Aβ1-42, and (2) hippocampal slices superfused with Aβ1-42 peptides. In hippocampal gliosomes of APP/PS1 mice, Cx43 levels were increased, whereas mice administered icv with Aβ1-42 only displayed increased Cx43 phosphorylation levels. This suggests that hemichannel activity might be differentially modulated throughout AD progression. Additionally, we tested if adenosine A2A receptor (A2AR) blockade reversed alterations of astrocytic hemichannel activity and found that the pharmacological blockade or genetic silencing (global and astrocytic) of A2AR prevented Aβ-induced hemichannel dysregulation in hippocampal slices, although A2AR genetic silencing increased the activity of astroglial hemichannels in control conditions. In primary cultures of astrocytes, A2AR-related protective effect was shown to occur through a protein kinase C (PKC) pathway. Our results indicate that the dysfunction of hemichannel activity in hippocampal astrocytes is an early event in AD, which is modulated by A2AR.
Collapse
Affiliation(s)
- Daniela Madeira
- Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Rua Larga, Polo I FMUC, First Floor, 3004-504, Coimbra, Portugal
| | - Joana Domingues
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Rua Larga, Polo I FMUC, First Floor, 3004-504, Coimbra, Portugal
| | - Cátia R Lopes
- Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Rua Larga, Polo I FMUC, First Floor, 3004-504, Coimbra, Portugal
| | - Paula M Canas
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Rua Larga, Polo I FMUC, First Floor, 3004-504, Coimbra, Portugal
| | - Rodrigo A Cunha
- Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Rua Larga, Polo I FMUC, First Floor, 3004-504, Coimbra, Portugal
| | - Paula Agostinho
- Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal.
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Rua Larga, Polo I FMUC, First Floor, 3004-504, Coimbra, Portugal.
| |
Collapse
|
33
|
Zhang L, Jia Z, Wu Q, Bai T, Wang B, Hu X, Li T, Liu X, Fu J, Chen Y, Ding X, Liu Z, Xu Z, Zhou H. Alleviating symptoms of neurodegenerative disorders by astrocyte-specific overexpression of TMEM164 in mice. Nat Metab 2023; 5:1787-1802. [PMID: 37679556 DOI: 10.1038/s42255-023-00887-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 08/08/2023] [Indexed: 09/09/2023]
Abstract
Neuroinflammatory microglia secrete cytokines to induce neurotoxic reactive astrocytes, which are one of the major causes of neuronal death. However, the intrinsic key regulators underlying neurotoxic reactive astrocytes induction are unknown. Here we show that the transmembrane protein 164 (TMEM164) is an early-response intrinsic factor that regulates neurotoxic astrocyte reactivity. TMEM164 overexpression inhibits the induction of neurotoxic reactive astrocytes, maintains normal astrocytic functions and suppresses neurotoxic reactive astrocyte-mediated neuronal death by decreasing the secretion of neurotoxic saturated lipids. Adeno-associated virus-mediated, astrocyte-specific TMEM164 overexpression in male and female mice prevents the induction of neurotoxic reactive astrocytes, dopaminergic neuronal loss and motor deficits in a Parkinson's disease model. Notably, brain-wide astrocyte-specific TMEM164 overexpression prevents the induction of neurotoxic reactive astrocytes, amyloid β deposition, neurodegeneration and memory decline in the 5XFAD Alzheimer's disease mouse model, suggesting that TMEM164 could serve as a potential therapeutic target for neurodegenerative disorders.
Collapse
Affiliation(s)
- Liansheng Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Zhiheng Jia
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Qiang Wu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Tao Bai
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Bo Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xinde Hu
- Genemagic Biosciences, Shanghai, China
| | - Tianwen Li
- Fudan University Huashan Hospital, Department of Neurosurgery, National Center for Neurological Disorders, National Key Laboratory for Medical Neurobiology, Shanghai Key Library of Brain Function and Regeneration, Institutes of Brain Science, MOE Frontiers Center for Brain Science, Shanghai Medical College-Fudan University, Shanghai, China
| | - Xingyu Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Jiqiang Fu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yuelei Chen
- Stem Cell Bank/Stem Cell Core Facility, Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoyan Ding
- Stem Cell Bank/Stem Cell Core Facility, Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Zhen Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Zhengzheng Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| | - Haibo Zhou
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China.
| |
Collapse
|
34
|
Kim MS, Cho K, Cho MH, Kim NY, Kim K, Kim DH, Yoon SY. Neuronal MHC-I complex is destabilized by amyloid-β and its implications in Alzheimer's disease. Cell Biosci 2023; 13:181. [PMID: 37773139 PMCID: PMC10540404 DOI: 10.1186/s13578-023-01132-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 09/11/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUNDS The expression of major histocompatibility complex I (MHC-I) in neurons has recently been shown to regulate neurite outgrowth and synaptic plasticity. However, its contribution to neurodegenerative diseases such as Alzheimer's disease (AD) remains largely unknown. METHODS In this study, we investigated the relationship between impaired MHC-I-β2M complex and AD in vitro and human AD samples. Interaction between protein was identified by liquid chromatography-tandem mass spectrometry and confirmed by immunoprecipitation. Single-chain trimer of MHC-I-β2M was generated to study the effect of stabilization of MHC-I-β2M complex on NCAM1 signaling. RESULTS MHC-I is destabilized in the brains of AD patients and neuronal cells treated with oligomeric β-amyloid (Aβ). Specifically, Aβ oligomers disassemble the MHC-I-β2-microglobulin (β2M) complex, leading to reduced interactions with neural cell adhesion molecule 1 (NCAM1), a novel interactor of neuronal MHC-I, and decreased signaling. Inhibition of MHC-I-β2M complex destabilization by non-dissociable MHC-I-β2M-peptide complex restored MHC-I-NCAM1 signaling in neuronal cells. CONCLUSIONS The current study demonstrated that disruption of MHC-1-NCAM1 signaling by Aβ induced disassembly of MHC-I-β2M complex is involved in the pathophysiology of AD. Moreover, our findings suggest modulation of MHC-I stability may be a potential therapeutic target for restoring synaptic function in AD.
Collapse
Affiliation(s)
- Min-Seok Kim
- ADEL Institute of Science & Technology (AIST), ADEL, Inc., Seoul, Korea
| | - Kwangmin Cho
- ADEL Institute of Science & Technology (AIST), ADEL, Inc., Seoul, Korea
| | - Mi-Hyang Cho
- ADEL Institute of Science & Technology (AIST), ADEL, Inc., Seoul, Korea
| | - Na-Young Kim
- ADEL Institute of Science & Technology (AIST), ADEL, Inc., Seoul, Korea
| | - Kyunggon Kim
- Department of Convergence Medicine, Convergence Medicine Research Center/Biomedical Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Dong-Hou Kim
- Department of Brain Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Seung-Yong Yoon
- ADEL Institute of Science & Technology (AIST), ADEL, Inc., Seoul, Korea.
- Department of Brain Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
35
|
Roggeri A, Olivero G, Usai C, Vanmierlo T, Pittaluga A. Presynaptic Release-Regulating Sphingosine 1-Phosphate 1/3 Receptors in Cortical Glutamatergic Terminals: Adaptations in EAE Mice and Impact of Therapeutic FTY720. Cells 2023; 12:2343. [PMID: 37830557 PMCID: PMC10571862 DOI: 10.3390/cells12192343] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/13/2023] [Accepted: 09/20/2023] [Indexed: 10/14/2023] Open
Abstract
This study provides evidence of the existence of presynaptic inhibitory sphingosine-1-phosphate receptor 1 (S1P1R) and facilitatory S1P3R in cortical nerve endings (synaptosomes) of healthy mice. The conclusion relies on the findings that (i) the S1P1R agonist CS-2100 (0.1-30 nM) inhibits the 12 mM KCl-evoked glutamate exocytosis (quantified as the release of [3H]D-aspartate) while the S1P3R allosteric agonist CYM-5541 potentiates it and (ii) these effects are inhibited by the S1P1R antagonist Ex 26 (30-300 nM) and the S1P3R antagonist TY-52156 (100-1000 nM), respectively. Confocal microscopy and western blot analysis confirmed the presence of S1P1R and S1P3R proteins in cortical glutamatergic synaptosomes, which were scarcely accessible to biotin in a biotinylation study. Then, we demonstrated that S1P1R and S1P3R densities and their release activity are amplified in cortical synaptosomes of mice suffering from experimental autoimmune encephalomyelitis (EAE), despite receptors maintain their preferential internal distribution. Receptor changes recover following chronic oral therapeutic FTY720 (0.03 mg/Kg/day). These results improve our knowledge of the role of presynaptic release-regulating S1P1Rs and S1P3Rs controlling glutamate transmission in the CNS also unravelling functional adaptations during EAE that recover following chronic FTY720. In a whole, these findings provide new information on the central neuroprotectant activities of FTY720.
Collapse
Affiliation(s)
- Alessandra Roggeri
- Department of Pharmacy (DiFar), University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy; (A.R.); (G.O.)
| | - Guendalina Olivero
- Department of Pharmacy (DiFar), University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy; (A.R.); (G.O.)
| | - Cesare Usai
- Institute of Biophysics, National Research Council, Via De Marini 6, 16149 Genoa, Italy;
| | - Tim Vanmierlo
- Department of Neuroscience, Biomedical Research Institute, European Graduate School of Neuroscience, Hasselt University, B-3590 Hasselt, Belgium;
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neurosciences, Division Translational Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Anna Pittaluga
- Department of Pharmacy (DiFar), Center of Excellence for Biomedical Research, 3Rs Center, University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, 16145 Genoa, Italy
| |
Collapse
|
36
|
Byun YG, Kim NS, Kim G, Jeon YS, Choi JB, Park CW, Kim K, Jang H, Kim J, Kim E, Han YM, Yoon KJ, Lee SH, Chung WS. Stress induces behavioral abnormalities by increasing expression of phagocytic receptor MERTK in astrocytes to promote synapse phagocytosis. Immunity 2023; 56:2105-2120.e13. [PMID: 37527657 DOI: 10.1016/j.immuni.2023.07.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 05/09/2023] [Accepted: 07/06/2023] [Indexed: 08/03/2023]
Abstract
Childhood neglect and/or abuse can induce mental health conditions with unknown mechanisms. Here, we identified stress hormones as strong inducers of astrocyte-mediated synapse phagocytosis. Using in vitro, in vivo, and human brain organoid experiments, we showed that stress hormones increased the expression of the Mertk phagocytic receptor in astrocytes through glucocorticoid receptor (GR). In post-natal mice, exposure to early social deprivation (ESD) specifically activated the GR-MERTK pathway in astrocytes, but not in microglia. The excitatory post-synaptic density in cortical regions was reduced in ESD mice, and there was an increase in the astrocytic engulfment of these synapses. The loss of excitatory synapses, abnormal neuronal network activities, and behavioral abnormalities in ESD mice were largely prevented by ablating GR or MERTK in astrocytes. Our work reveals the critical roles of astrocytic GR-MERTK activation in evoking stress-induced abnormal behaviors in mice, suggesting GR-MERTK signaling as a therapeutic target for stress-induced mental health conditions.
Collapse
Affiliation(s)
- Youkyeong Gloria Byun
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Nam-Shik Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Gyuri Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Yi-Seon Jeon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jong Bin Choi
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Chan-Woo Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Kyungdeok Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Hyunsoo Jang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jinkyeong Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Yong-Mahn Han
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Ki-Jun Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Seung-Hee Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Won-Suk Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea.
| |
Collapse
|
37
|
Wang Y, Zhu Y, Li W, Yan S, Li C, Ma K, Hu M, Du C, Fu L, Sun J, Zhang CX. Synaptotagmin-11 Inhibits Synaptic Vesicle Endocytosis via Endophilin A1. J Neurosci 2023; 43:6230-6248. [PMID: 37474308 PMCID: PMC10490507 DOI: 10.1523/jneurosci.1348-21.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 05/12/2023] [Accepted: 06/02/2023] [Indexed: 07/22/2023] Open
Abstract
Synaptic vesicle (SV) endocytosis is a critical and well-regulated process for the maintenance of neurotransmission. We previously reported that synaptotagmin-11 (Syt11), an essential non-Ca2+-binding Syt associated with brain diseases, inhibits neuronal endocytosis (Wang et al., 2016). Here, we found that Syt11 deficiency caused accelerated SV endocytosis and vesicle recycling under sustained stimulation and led to the abnormal membrane partition of synaptic proteins in mouse hippocampal boutons of either sex. Furthermore, our study revealed that Syt11 has direct but Ca2+-independent binding with endophilin A1 (EndoA1), a membrane curvature sensor and endocytic protein recruiter, with high affinity. EndoA1-knockdown significantly reversed Syt11-KO phenotype, identifying EndoA1 as a main inhibitory target of Syt11 during SV endocytosis. The N-terminus of EndoA1 and the C2B domain of Syt11 were responsible for this interaction. A peptide (amino acids 314-336) derived from the Syt11 C2B efficiently blocked Syt11-EndoA1 binding both in vitro and in vivo Application of this peptide inhibited SV endocytosis in WT hippocampal neurons but not in EndoA1-knockdown neurons. Moreover, intracellular application of this peptide in mouse calyx of Held terminals of either sex effectively hampered both fast and slow SV endocytosis at physiological temperature. We thus propose that Syt11 ensures the precision of protein retrieval during SV endocytosis by inhibiting EndoA1 function at neuronal terminals.SIGNIFICANCE STATEMENT Endocytosis is a key stage of synaptic vesicle (SV) recycling. SV endocytosis retrieves vesicular membrane and protein components precisely to support sustained neurotransmission. However, the molecular mechanisms underlying the regulation of SV endocytosis remain elusive. Here, we reported that Syt11-KO accelerated SV endocytosis and impaired membrane partition of synaptic proteins. EndoA1 was identified as a main inhibitory target of Syt11 during SV endocytosis. Our study reveals a novel inhibitory mechanism of SV endocytosis in preventing hyperactivation of endocytosis, potentially safeguarding the recycling of synaptic proteins during sustained neurotransmission.
Collapse
Affiliation(s)
- Yalong Wang
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences; CAS Key Laboratory of Brain Connectome and Manipulation; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong 518055, China
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Ying Zhu
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wanru Li
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Shuxin Yan
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Chao Li
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Kunpeng Ma
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences; CAS Key Laboratory of Brain Connectome and Manipulation; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong 518055, China
| | - Meiqin Hu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Cuilian Du
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Lei Fu
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China
| | - Jianyuan Sun
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences; CAS Key Laboratory of Brain Connectome and Manipulation; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong 518055, China
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Claire Xi Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
38
|
Underwood EL, Redell JB, Hood KN, Maynard ME, Hylin M, Waxham MN, Zhao J, Moore AN, Dash PK. Enhanced presynaptic mitochondrial energy production is required for memory formation. Sci Rep 2023; 13:14431. [PMID: 37660191 PMCID: PMC10475119 DOI: 10.1038/s41598-023-40877-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 08/17/2023] [Indexed: 09/04/2023] Open
Abstract
Some of the prominent features of long-term memory formation include protein synthesis, gene expression, enhanced neurotransmitter release, increased excitability, and formation of new synapses. As these processes are critically dependent on mitochondrial function, we hypothesized that increased mitochondrial respiration and dynamics would play a prominent role in memory formation. To address this possibility, we measured mitochondrial oxygen consumption (OCR) in hippocampal tissue punches from trained and untrained animals. Our results show that context fear training significantly increased basal, ATP synthesis-linked, and maximal OCR in the Shaffer collateral-CA1 synaptic region, but not in the CA1 cell body layer. These changes were recapitulated in synaptosomes isolated from the hippocampi of fear-trained animals. As dynamin-related protein 1 (Drp1) plays an important role in mitochondrial fission, we examined its role in the increased mitochondrial respiration observed after fear training. Drp1 inhibitors decreased the training-associated enhancement of OCR and impaired contextual fear memory, but did not alter the number of synaptosomes containing mitochondria. Taken together, our results show context fear training increases presynaptic mitochondria respiration, and that Drp-1 mediated enhanced energy production in CA1 pre-synaptic terminals is necessary for context fear memory that does not result from an increase in the number of synaptosomes containing mitochondria or an increase in mitochondrial mass within the synaptic layer.
Collapse
Affiliation(s)
- Erica L Underwood
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, P.O. Box 20708, Houston, TX, 77225, USA
| | - John B Redell
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, P.O. Box 20708, Houston, TX, 77225, USA.
| | - Kimberly N Hood
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, P.O. Box 20708, Houston, TX, 77225, USA
| | - Mark E Maynard
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, P.O. Box 20708, Houston, TX, 77225, USA
- Department of Electrical and Computer Engineering, University of Houston, Houston, TX, USA
| | - Michael Hylin
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, P.O. Box 20708, Houston, TX, 77225, USA
- Department of Psychology, Southern Illinois University, Carbondale, IL, USA
| | - M Neal Waxham
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, P.O. Box 20708, Houston, TX, 77225, USA
| | - Jing Zhao
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, P.O. Box 20708, Houston, TX, 77225, USA
| | - Anthony N Moore
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, P.O. Box 20708, Houston, TX, 77225, USA
| | - Pramod K Dash
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, P.O. Box 20708, Houston, TX, 77225, USA
| |
Collapse
|
39
|
Wang Z, Xu Z, Luo Y, Peng S, Song H, Li T, Zheng J, Liu N, Wu S, Zhang J, Zhang L, Hu Y, Liu Y, Lu D, Dai J, Zhang J. Reduced biophotonic activities and spectral blueshift in Alzheimer's disease and vascular dementia models with cognitive impairment. Front Aging Neurosci 2023; 15:1208274. [PMID: 37727319 PMCID: PMC10505668 DOI: 10.3389/fnagi.2023.1208274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 08/18/2023] [Indexed: 09/21/2023] Open
Abstract
Background Although clinically, Alzheimer's disease (AD) and vascular dementia (VaD) are the two major types of dementia, it is unclear whether the biophotonic activities associated with cognitive impairments in these diseases share common pathological features. Methods We used the ultraweak biophoton imaging system (UBIS) and synaptosomes prepared by modified percoll method to directly evaluate the functional changes in synapses and neural circuits in AD and VaD model animals. Results We found that biophotonic activities induced by glutamate were significantly reduced and spectral blueshifted in synaptosomes and brain slices. These changes could be partially reversed by pre-perfusion of the ifenprodil, a specific antagonist of the GluN2B subunit of N-methyl-D-aspartate receptors (NMDARs). Conclusion Our findings suggest that AD and VaD pathology present similar but complex changes in biophotonic activities and transmission at synapses and neural circuits, implying that communications and information processing of biophotonic signals in the brain are crucial for advanced cognitive functions.
Collapse
Affiliation(s)
- Zhuo Wang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
- College of Life Science, Wuhan Institute for Neuroscience and Neuroengineering, South-Central Minzu University, Wuhan, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhipeng Xu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yi Luo
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Sisi Peng
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hao Song
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Tian Li
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiaxin Zheng
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Na Liu
- College of Life Science, Wuhan Institute for Neuroscience and Neuroengineering, South-Central Minzu University, Wuhan, China
| | - Shenjia Wu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Junxia Zhang
- Academy of Chinese Medical Sciences, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Lei Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuan Hu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yanping Liu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Dongwei Lu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiapei Dai
- College of Life Science, Wuhan Institute for Neuroscience and Neuroengineering, South-Central Minzu University, Wuhan, China
| | - Junjian Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
40
|
Odell LR, Jones NC, Chau N, Robertson MJ, Ambrus JI, Deane FM, Young KA, Whiting A, Xue J, Prichard K, Daniel JA, Gorgani NN, O'Brien TJ, Robinson PJ, McCluskey A. The sulfonadyns: a class of aryl sulfonamides inhibiting dynamin I GTPase and clathrin mediated endocytosis are anti-seizure in animal models. RSC Med Chem 2023; 14:1492-1511. [PMID: 37593570 PMCID: PMC10429932 DOI: 10.1039/d2md00371f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 04/15/2023] [Indexed: 08/19/2023] Open
Abstract
We show that dansylcadaverine (1) a known in-cell inhibitor of clathrin mediated endocytosis (CME), moderately inhibits dynamin I (dynI) GTPase activity (IC50 45 μM) and transferrin (Tfn) endocytosis in U2OS cells (IC50 205 μM). Synthesis gave a new class of GTP-competitive dynamin inhibitors, the Sulfonadyns™. The introduction of a terminal cinnamyl moiety greatly enhanced dynI inhibition. Rigid diamine or amide links between the dansyl and cinnamyl moieties were detrimental to dynI inhibition. Compounds with in vitro inhibition of dynI activity <10 μM were tested in-cell for inhibition of CME. These data unveiled a number of compounds, e.g. analogues 33 ((E)-N-(6-{[(3-(4-bromophenyl)-2-propen-1-yl]amino}hexyl)-5-isoquinolinesulfonamide)) and 47 ((E)-N-(3-{[3-(4-bromophenyl)-2-propen-1-yl]amino}propyl)-1-naphthalenesulfonamide)isomers that showed dyn IC50 <4 μM, IC50(CME) <30 μM and IC50(SVE) from 12-265 μM. Both analogues (33 and 47) are at least 10 times more potent that the initial lead, dansylcadaverine (1). Enzyme kinetics revealed these sulfonamide analogues as being GTP competitive inhibitors of dynI. Sulfonadyn-47, the most potent SVE inhibitor observed (IC50(SVE) = 12.3 μM), significantly increased seizure threshold in a 6 Hz mouse psychomotor seizure test at 30 (p = 0.003) and 100 mg kg-1 ip (p < 0.0001), with similar anti-seizure efficacy to the established anti-seizure medication, sodium valproate (400 mg kg-1). The Sulfonadyn™ class of drugs target dynamin and show promise as novel leads for future anti-seizure medications.
Collapse
Affiliation(s)
- Luke R Odell
- Chemistry, Centre for Chemical Biology, School of Environmental & Life Science, The University of Newcastle University Drive Callaghan NSW 2308 Australia +612 4921 5472 +612 4921 6486
| | - Nigel C Jones
- Department of Neuroscience, Central Clinical School, Monash University Melbourne Victoria 3004 Australia
- Department of Neurology, The Alfred Hospital Commercial Road Melbourne Victoria 3004 Australia
- Department of Medicine (Royal Melbourne Hospital), University of Melbourne Parkville Victoria 3052 Australia
| | - Ngoc Chau
- Cell Signaling Unit, Children's Medical Research Institute, The University of Sydney 214 Hawkesbury Road Westmead NSW 2145 Australia +612 8865 2915
| | - Mark J Robertson
- Chemistry, Centre for Chemical Biology, School of Environmental & Life Science, The University of Newcastle University Drive Callaghan NSW 2308 Australia +612 4921 5472 +612 4921 6486
| | - Joseph I Ambrus
- Chemistry, Centre for Chemical Biology, School of Environmental & Life Science, The University of Newcastle University Drive Callaghan NSW 2308 Australia +612 4921 5472 +612 4921 6486
| | - Fiona M Deane
- Chemistry, Centre for Chemical Biology, School of Environmental & Life Science, The University of Newcastle University Drive Callaghan NSW 2308 Australia +612 4921 5472 +612 4921 6486
| | - Kelly A Young
- Chemistry, Centre for Chemical Biology, School of Environmental & Life Science, The University of Newcastle University Drive Callaghan NSW 2308 Australia +612 4921 5472 +612 4921 6486
| | - Ainslie Whiting
- Cell Signaling Unit, Children's Medical Research Institute, The University of Sydney 214 Hawkesbury Road Westmead NSW 2145 Australia +612 8865 2915
| | - Jing Xue
- Cell Signaling Unit, Children's Medical Research Institute, The University of Sydney 214 Hawkesbury Road Westmead NSW 2145 Australia +612 8865 2915
| | - Kate Prichard
- Chemistry, Centre for Chemical Biology, School of Environmental & Life Science, The University of Newcastle University Drive Callaghan NSW 2308 Australia +612 4921 5472 +612 4921 6486
| | - James A Daniel
- Cell Signaling Unit, Children's Medical Research Institute, The University of Sydney 214 Hawkesbury Road Westmead NSW 2145 Australia +612 8865 2915
| | - Nick N Gorgani
- Cell Signaling Unit, Children's Medical Research Institute, The University of Sydney 214 Hawkesbury Road Westmead NSW 2145 Australia +612 8865 2915
| | - Terence J O'Brien
- Department of Neurology, The Alfred Hospital Commercial Road Melbourne Victoria 3004 Australia
- Department of Medicine (Royal Melbourne Hospital), University of Melbourne Parkville Victoria 3052 Australia
| | - Phillip J Robinson
- Cell Signaling Unit, Children's Medical Research Institute, The University of Sydney 214 Hawkesbury Road Westmead NSW 2145 Australia +612 8865 2915
| | - Adam McCluskey
- Chemistry, Centre for Chemical Biology, School of Environmental & Life Science, The University of Newcastle University Drive Callaghan NSW 2308 Australia +612 4921 5472 +612 4921 6486
| |
Collapse
|
41
|
Lichter EZ, Trease AJ, Cooper K, Stauch KL, Fox HS. Effects of Parkin on the Mitochondrial Genome in the Heart and Brain of Mitochondrial Mutator Mice. Adv Biol (Weinh) 2023; 7:e2300154. [PMID: 37376822 DOI: 10.1002/adbi.202300154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Indexed: 06/29/2023]
Abstract
Mitochondrial dysfunction has been implicated in neurodegenerative diseases like Parkinson's disease (PD). This study investigates the role of Parkin, a protein involved in mitochondrial quality control, and strongly linked to PD, in the context of mitochondrial DNA (mtDNA) mutations. Mitochondrial mutator mice (PolgD257A/D257A ) (Polg) are used and bred with Parkin knockout (PKO) mice or mice with disinhibited Parkin (W402A). In the brain, mtDNA mutations are analyzed in synaptosomes, presynaptic neuronal terminals, which are far from neuronal soma, which likely renders mitochondria there more vulnerable compared with brain homogenate. Surprisingly, PKO results in reduced mtDNA mutations in the brain but increased control region multimer (CRM) in synaptosomes. In the heart, both PKO and W402A lead to increased mutations, with W402A showing more mutations in the heart than PKO. Computational analysis reveals many of these mutations are deleterious. These findings suggest that Parkin plays a tissue-dependent role in regulating mtDNA damage response, with differential effects in the brain and heart. Understanding the specific role of Parkin in different tissues may provide insights into the underlying mechanisms of PD and potential therapeutic strategies. Further investigation into these pathways can enhance the understanding of neurodegenerative diseases associated with mitochondrial dysfunction.
Collapse
Affiliation(s)
- Eliezer Z Lichter
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Andrew J Trease
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kathryn Cooper
- School of Interdisciplinary Informatics, University of Nebraska Omaha, Omaha, NE, 68182, USA
| | - Kelly L Stauch
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Howard S Fox
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| |
Collapse
|
42
|
Papantoniou C, Laugks U, Betzin J, Capitanio C, Ferrero JJ, Sánchez-Prieto J, Schoch S, Brose N, Baumeister W, Cooper BH, Imig C, Lučić V. Munc13- and SNAP25-dependent molecular bridges play a key role in synaptic vesicle priming. SCIENCE ADVANCES 2023; 9:eadf6222. [PMID: 37343100 PMCID: PMC10284560 DOI: 10.1126/sciadv.adf6222] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 05/17/2023] [Indexed: 06/23/2023]
Abstract
Synaptic vesicle tethering, priming, and neurotransmitter release require a coordinated action of multiple protein complexes. While physiological experiments, interaction data, and structural studies of purified systems were essential for our understanding of the function of the individual complexes involved, they cannot resolve how the actions of individual complexes integrate. We used cryo-electron tomography to simultaneously image multiple presynaptic protein complexes and lipids at molecular resolution in their native composition, conformation, and environment. Our detailed morphological characterization suggests that sequential synaptic vesicle states precede neurotransmitter release, where Munc13-comprising bridges localize vesicles <10 nanometers and soluble N-ethylmaleimide-sensitive factor attachment protein 25-comprising bridges <5 nanometers from the plasma membrane, the latter constituting a molecularly primed state. Munc13 activation supports the transition to the primed state via vesicle bridges to plasma membrane (tethers), while protein kinase C promotes the same transition by reducing vesicle interlinking. These findings exemplify a cellular function performed by an extended assembly comprising multiple molecularly diverse complexes.
Collapse
Affiliation(s)
- Christos Papantoniou
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Ulrike Laugks
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Julia Betzin
- Department of Neuropathology, University Hospital of Bonn, 53127 Bonn, Germany
| | - Cristina Capitanio
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - José Javier Ferrero
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Universidad Complutense, and Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - José Sánchez-Prieto
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Universidad Complutense, and Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Susanne Schoch
- Department of Neuropathology, University Hospital of Bonn, 53127 Bonn, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute of Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany
| | - Wolfgang Baumeister
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Benjamin H. Cooper
- Department of Molecular Neurobiology, Max Planck Institute of Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany
| | - Cordelia Imig
- Department of Molecular Neurobiology, Max Planck Institute of Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany
- Department of Neuroscience, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Vladan Lučić
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| |
Collapse
|
43
|
Yim YY, Nestler EJ. Cell-Type-Specific Neuroproteomics of Synapses. Biomolecules 2023; 13:998. [PMID: 37371578 PMCID: PMC10296650 DOI: 10.3390/biom13060998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/08/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
In the last two decades, our knowledge of synaptic proteomes and their relationship to normal brain function and neuropsychiatric disorders has been expanding rapidly through the use of more powerful neuroproteomic approaches. However, mass spectrometry (MS)-based neuroproteomic studies of synapses still require cell-type, spatial, and temporal proteome information. With the advancement of sample preparation and MS techniques, we have just begun to identify and understand proteomes within a given cell type, subcellular compartment, and cell-type-specific synapse. Here, we review the progress and limitations of MS-based neuroproteomics of synapses in the mammalian CNS and highlight the recent applications of these approaches in studying neuropsychiatric disorders such as major depressive disorder and substance use disorders. Combining neuroproteomic findings with other omics studies can generate an in-depth, comprehensive map of synaptic proteomes and possibly identify new therapeutic targets and biomarkers for several central nervous system disorders.
Collapse
Affiliation(s)
- Yun Young Yim
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | | |
Collapse
|
44
|
Yokozeki T, Hama Y, Fujita K, Igarashi T, Hirama M, Tsumuraya T. Evaluation of relative potency of calibrated ciguatoxin congeners by near-infrared fluorescent receptor binding and neuroblastoma cell-based assays. Toxicon 2023; 230:107161. [PMID: 37201801 DOI: 10.1016/j.toxicon.2023.107161] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/08/2023] [Accepted: 05/15/2023] [Indexed: 05/20/2023]
Abstract
Ciguatera fish poisoning (CFP) is a foodborne illness affecting > 50,000 people worldwide annually. It is caused by eating marine invertebrates and fish that have accumulated ciguatoxins (CTXs). Recently, the risk of CFP to human health, the local economy, and fishery resources have increased; therefore, detection methods are urgently needed. Functional assays for detecting ciguatoxins in fish include receptor binding (RBA) and neuroblastoma cell-based assay (N2a assay), which can detect all CTX congeners. In this study, we made these assays easier to use. For RBA, an assay was developed using a novel near-infrared fluorescent ligand, PREX710-BTX, to save valuable CTXs. In the N2a assay, a 1-day assay was developed with the same detection performance as the conventional 2-day assay. Additionally, in these assays, we used calibrated CTX standards from the Pacific determined by quantitative NMR for the first time to compare the relative potency of congeners, which differed significantly among previous studies. In the RBA, there was almost no difference in the binding affinity among congeners, showing that the differences in side chains, stereochemistry, and backbone structure of CTXs did not affect the binding affinity. However, this result did not correlate with the toxic equivalency factors (TEFs) based on acute toxicity in mice. In contrast, the N2a assay showed a good correlation with TEFs based on acute toxicity in mice, except for CTX3C. These findings, obtained with calibrated toxin standards, provide important insights into evaluating the total toxicity of CTXs using functional assays.
Collapse
Affiliation(s)
- Toshiaki Yokozeki
- Japan Food Research Laboratories, Osaka Saito Laboratory, 7-4-41 Saitoasagi, Ibaraki shi, Osaka, 567-0085, Japan; Osaka Prefecture University, Department of Biological Science, Graduate School of Science, 1-2 Gakuen-cho, Sakai-shi, Osaka, 599-8570, Japan; Osaka Metropolitan University, Department of Biological Chemistry, Graduate School of Science, 1-2 Gakuen-cho, Sakai-shi, Osaka, 599-8570, Japan
| | - Yuka Hama
- Osaka Prefecture University, Department of Biological Science, Graduate School of Science, 1-2 Gakuen-cho, Sakai-shi, Osaka, 599-8570, Japan
| | - Kazuhiro Fujita
- Japan Food Research Laboratories, Osaka Saito Laboratory, 7-4-41 Saitoasagi, Ibaraki shi, Osaka, 567-0085, Japan
| | - Tomoji Igarashi
- Japan Food Research Laboratories, Tama Laboratory, 6-11-10 Nagayama, Tama-shi, Tokyo, 206-0025, Japan
| | - Masahiro Hirama
- Osaka Prefecture University, Department of Biological Science, Graduate School of Science, 1-2 Gakuen-cho, Sakai-shi, Osaka, 599-8570, Japan; Osaka Metropolitan University, Department of Biological Chemistry, Graduate School of Science, 1-2 Gakuen-cho, Sakai-shi, Osaka, 599-8570, Japan
| | - Takeshi Tsumuraya
- Osaka Prefecture University, Department of Biological Science, Graduate School of Science, 1-2 Gakuen-cho, Sakai-shi, Osaka, 599-8570, Japan; Osaka Metropolitan University, Department of Biological Chemistry, Graduate School of Science, 1-2 Gakuen-cho, Sakai-shi, Osaka, 599-8570, Japan.
| |
Collapse
|
45
|
Gorski K, Jackson CB, Nyman TA, Rezov V, Battersby BJ, Lehesjoki AE. Progressive mitochondrial dysfunction in cerebellar synaptosomes of cystatin B-deficient mice. Front Mol Neurosci 2023; 16:1175851. [PMID: 37251643 PMCID: PMC10213208 DOI: 10.3389/fnmol.2023.1175851] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/25/2023] [Indexed: 05/31/2023] Open
Abstract
The involvement of mitochondrial dysfunction in cystatin B (CSTB) deficiency has been suggested, but its role in the onset of neurodegeneration, myoclonus, and ataxia in the CSTB-deficient mouse model (Cstb-/-) is yet unknown. CSTB is an inhibitor of lysosomal and nuclear cysteine cathepsins. In humans, partial loss-of-function mutations cause the progressive myoclonus epilepsy neurodegenerative disorder, EPM1. Here we applied proteome analysis and respirometry on cerebellar synaptosomes from early symptomatic (Cstb-/-) mice to identify the molecular mechanisms involved in the onset of CSTB-deficiency associated neural pathogenesis. Proteome analysis showed that CSTB deficiency is associated with differential expression of mitochondrial and synaptic proteins, and respirometry revealed a progressive impairment in mitochondrial function coinciding with the onset of myoclonus and neurodegeneration in (Cstb-/-) mice. This mitochondrial dysfunction was not associated with alterations in mitochondrial DNA copy number or membrane ultrastructure. Collectively, our results show that CSTB deficiency generates a defect in synaptic mitochondrial bioenergetics that coincides with the onset and progression of the clinical phenotypes, and thus is likely a contributor to the pathogenesis of EPM1.
Collapse
Affiliation(s)
- Katarin Gorski
- Folkhälsan Research Center, Helsinki, Finland
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Christopher B. Jackson
- Department of Biochemistry and Developmental Biology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tuula A. Nyman
- Department of Immunology, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Veronika Rezov
- Folkhälsan Research Center, Helsinki, Finland
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | | - Anna-Elina Lehesjoki
- Folkhälsan Research Center, Helsinki, Finland
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
46
|
Radecke J, Seeger R, Kádková A, Laugks U, Khosrozadeh A, Goldie KN, Lučić V, Sørensen JB, Zuber B. Morphofunctional changes at the active zone during synaptic vesicle exocytosis. EMBO Rep 2023; 24:e55719. [PMID: 36876590 PMCID: PMC10157379 DOI: 10.15252/embr.202255719] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 01/30/2023] [Accepted: 02/16/2023] [Indexed: 03/07/2023] Open
Abstract
Synaptic vesicle (SV) fusion with the plasma membrane (PM) proceeds through intermediate steps that remain poorly resolved. The effect of persistent high or low exocytosis activity on intermediate steps remains unknown. Using spray-mixing plunge-freezing cryo-electron tomography we observe events following synaptic stimulation at nanometer resolution in near-native samples. Our data suggest that during the stage that immediately follows stimulation, termed early fusion, PM and SV membrane curvature changes to establish a point contact. The next stage-late fusion-shows fusion pore opening and SV collapse. During early fusion, proximal tethered SVs form additional tethers with the PM and increase the inter-SV connector number. In the late-fusion stage, PM-proximal SVs lose their interconnections, allowing them to move toward the PM. Two SNAP-25 mutations, one arresting and one disinhibiting spontaneous release, cause connector loss. The disinhibiting mutation causes loss of membrane-proximal multiple-tethered SVs. Overall, tether formation and connector dissolution are triggered by stimulation and respond to spontaneous fusion rate manipulation. These morphological observations likely correspond to SV transition from one functional pool to another.
Collapse
Affiliation(s)
- Julika Radecke
- Institute of AnatomyUniversity of BernBernSwitzerland
- Department of Neuroscience, University of CopenhagenCopenhagenDenmark
- Diamond Light Source LtdDidcotUK
- Graduate School for Cellular and Biomedical SciencesUniversity of BernBernSwitzerland
| | - Raphaela Seeger
- Institute of AnatomyUniversity of BernBernSwitzerland
- Graduate School for Cellular and Biomedical SciencesUniversity of BernBernSwitzerland
| | - Anna Kádková
- Department of Neuroscience, University of CopenhagenCopenhagenDenmark
| | - Ulrike Laugks
- Max‐Planck‐Institute of BiochemistryMartinsriedGermany
| | - Amin Khosrozadeh
- Institute of AnatomyUniversity of BernBernSwitzerland
- Graduate School for Cellular and Biomedical SciencesUniversity of BernBernSwitzerland
| | | | - Vladan Lučić
- Max‐Planck‐Institute of BiochemistryMartinsriedGermany
| | - Jakob B Sørensen
- Department of Neuroscience, University of CopenhagenCopenhagenDenmark
| | - Benoît Zuber
- Institute of AnatomyUniversity of BernBernSwitzerland
| |
Collapse
|
47
|
Hsu SK, Lu CW, Chiu KM, Lee MY, Lin TY, Wang SJ. Mangiferin depresses vesicular glutamate release in synaptosomes from the rat cerebral cortex by decreasing synapsin I phosphorylation. Eur J Pharmacol 2023; 950:175772. [PMID: 37146708 DOI: 10.1016/j.ejphar.2023.175772] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/07/2023]
Abstract
Mangiferin is a glucosyl xanthone that has been shown to be a neuroprotective agent against brain disorders involving excess glutamate. However, the effect of mangiferin on the function of the glutamatergic system has not been investigated. In this study, we used synaptosomes from the rat cerebral cortex to investigate the effect of mangiferin on glutamate release and identify the possible underlying mechanism. We observed that mangiferin produced a concentration-dependent reduction in the release of glutamate elicited by 4-aminopyridine with an IC50 value of 25 μM. Inhibition of glutamate release was blocked by removing extracellular calcium and by treatment with the vacuolar-type H+-ATPase inhibitor bafilomycin A1, which prevents the uptake and storage of glutamate in vesicles. Moreover, we showed that mangiferin decreased the 4-aminopyridine-elicited FM1-43 release and synaptotagmin 1 luminal domain antibody (syt1-L ab) uptake from synaptosomes, which correlated with decreased synaptic vesicle exocytosis. Transmission electron microscopy in synaptosomes also showed that mangiferin attenuated the 4-aminopyridine-elicited decrease in the number of synaptic vesicles. In addition, antagonism of Ca2+/calmodulin-dependent kinase II (CaMKII) and protein kinase A (PKA) counteracted mangiferin's effect on glutamate release. Mangiferin also decreased the phosphorylation of CaMKII, PKA, and synapsin I elicited by 4-aminopyridine treatment. Our data suggest that mangiferin reduces PKA and CaMKII activation and synapsin I phosphorylation, which could decrease synaptic vesicle availability and lead to a subsequent reduction in vesicular glutamate release from synaptosomes.
Collapse
Affiliation(s)
- Szu-Kai Hsu
- School of Medicine, Fu Jen Catholic University, New Taipei City, 24205, Taiwan; Department of Neurosurgery, Cathay General Hospital, Taipei, 106438, Taiwan
| | - Cheng-Wei Lu
- Department of Anesthesiology, Far-Eastern Memorial Hospital, New Taipei City, 22060, Taiwan; Department of Mechanical Engineering, Yuan Ze University, Taoyuan, 32003, Taiwan
| | - Kuan-Ming Chiu
- Division of Cardiovascular Surgery, Cardiovascular Center, Far-Eastern Memorial Hospital, New Taipei City, 22060, Taiwan; Department of Electrical Engineering, Yuan Ze University, Taoyuan, 32003, Taiwan
| | - Ming-Yi Lee
- Department of Medical Research, Far-Eastern Memorial Hospital, New Taipei City, 22060, Taiwan
| | - Tzu-Yu Lin
- Department of Anesthesiology, Far-Eastern Memorial Hospital, New Taipei City, 22060, Taiwan; Department of Mechanical Engineering, Yuan Ze University, Taoyuan, 32003, Taiwan.
| | - Su-Jane Wang
- School of Medicine, Fu Jen Catholic University, New Taipei City, 24205, Taiwan; Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan City, 33303, Taiwan.
| |
Collapse
|
48
|
Popescu AS, Butler CA, Allendorf DH, Piers TM, Mallach A, Roewe J, Reinhardt P, Cinti A, Redaelli L, Boudesco C, Pradier L, Pocock JM, Thornton P, Brown GC. Alzheimer's disease-associated R47H TREM2 increases, but wild-type TREM2 decreases, microglial phagocytosis of synaptosomes and neuronal loss. Glia 2023; 71:974-990. [PMID: 36480007 PMCID: PMC10952257 DOI: 10.1002/glia.24318] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/22/2022] [Accepted: 11/25/2022] [Indexed: 12/14/2022]
Abstract
Triggering receptor on myeloid cells 2 (TREM2) is an innate immune receptor, upregulated on the surface of microglia associated with amyloid plaques in Alzheimer's disease (AD). Individuals heterozygous for the R47H variant of TREM2 have greatly increased risk of developing AD. We examined the effects of wild-type (WT), R47H and knock-out (KO) of human TREM2 expression in three microglial cell systems. Addition of mouse BV-2 microglia expressing R47H TREM2 to primary mouse neuronal cultures caused neuronal loss, not observed with WT TREM2. Neuronal loss was prevented by using annexin V to block exposed phosphatidylserine, an eat-me signal and ligand of TREM2, suggesting loss was mediated by microglial phagocytosis of neurons exposing phosphatidylserine. Addition of human CHME-3 microglia expressing R47H TREM2 to LUHMES neuronal-like cells also caused loss compared to WT TREM2. Expression of R47H TREM2 in BV-2 and CHME-3 microglia increased their uptake of phosphatidylserine-beads and synaptosomes versus WT TREM2. Human iPSC-derived microglia with heterozygous R47H TREM2 had increased phagocytosis of synaptosomes vs common-variant TREM2. Additionally, phosphatidylserine liposomes increased activation of human iPSC-derived microglia expressing homozygous R47H TREM2 versus common-variant TREM2. Finally, overexpression of TREM2 in CHME-3 microglia caused increased expression of cystatin F, a cysteine protease inhibitor, and knock-down of cystatin F increased CHME-3 uptake of phosphatidylserine-beads. Together, these data suggest that R47H TREM2 may increase AD risk by increasing phagocytosis of synapses and neurons via greater activation by phosphatidylserine and that WT TREM2 may decrease microglial phagocytosis of synapses and neurons via cystatin F.
Collapse
Affiliation(s)
- Alma S. Popescu
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| | - Claire A. Butler
- Department of BiochemistryUniversity of CambridgeCambridgeUK
- Neuroscience, BioPharmaceuticals R&DAstraZenecaCambridgeUK
| | | | - Thomas M. Piers
- Department of NeuroinflammationUCL Queen Square Institute of NeurologyLondonUK
| | - Anna Mallach
- Department of NeuroinflammationUCL Queen Square Institute of NeurologyLondonUK
| | - Julian Roewe
- Neuroscience DiscoveryAbbVie Deutschland GmbH & Co. KGLudwigshafenGermany
| | - Peter Reinhardt
- Neuroscience DiscoveryAbbVie Deutschland GmbH & Co. KGLudwigshafenGermany
| | | | | | | | | | - Jennifer M. Pocock
- Department of NeuroinflammationUCL Queen Square Institute of NeurologyLondonUK
| | - Peter Thornton
- Neuroscience, BioPharmaceuticals R&DAstraZenecaCambridgeUK
| | - Guy C. Brown
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| |
Collapse
|
49
|
Birkle TJY, Brown GC. Syk inhibitors protect against microglia-mediated neuronal loss in culture. Front Aging Neurosci 2023; 15:1120952. [PMID: 37009452 PMCID: PMC10050448 DOI: 10.3389/fnagi.2023.1120952] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 02/28/2023] [Indexed: 03/17/2023] Open
Abstract
Microglia are brain macrophages and play beneficial and/or detrimental roles in many brain pathologies because of their inflammatory and phagocytic activity. Microglial inflammation and phagocytosis are thought to be regulated by spleen tyrosine kinase (Syk), which is activated by multiple microglial receptors, including TREM2 (Triggering Receptor Expressed on Myeloid Cells 2), implicated in neurodegeneration. Here, we have tested whether Syk inhibitors can prevent microglia-dependent neurodegeneration induced by lipopolysaccharide (LPS) in primary neuron-glia cultures. We found that the Syk inhibitors BAY61-3606 and P505-15 (at 1 and 10 μM, respectively) completely prevented the neuronal loss induced by LPS, which was microglia-dependent. Syk inhibition also prevented the spontaneous loss of neurons from older neuron-glia cultures. In the absence of LPS, Syk inhibition depleted microglia from the cultures and induced some microglial death. However, in the presence of LPS, Syk inhibition had relatively little effect on microglial density (reduced by 0-30%) and opposing effects on the release of two pro-inflammatory cytokines (IL-6 decreased by about 45%, TNFα increased by 80%). Syk inhibition also had no effect on the morphological transition of microglia exposed to LPS. On the other hand, inhibition of Syk reduced microglial phagocytosis of beads, synapses and neurons. Thus, Syk inhibition in this model is most likely neuroprotective by reducing microglial phagocytosis, however, the reduced microglial density and IL-6 release may also contribute. This work adds to increasing evidence that Syk is a key regulator of the microglial contribution to neurodegenerative disease and suggests that Syk inhibitors may be used to prevent excessive microglial phagocytosis of synapses and neurons.
Collapse
Affiliation(s)
| | - Guy C. Brown
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
50
|
Dundee JM, Puigdellívol M, Butler R, Cockram TOJ, Brown GC. P2Y 6 receptor-dependent microglial phagocytosis of synapses mediates synaptic and memory loss in aging. Aging Cell 2022; 22:e13761. [PMID: 36565471 PMCID: PMC9924939 DOI: 10.1111/acel.13761] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/15/2022] [Accepted: 12/08/2022] [Indexed: 12/25/2022] Open
Abstract
Aging causes loss of brain synapses and memory, and microglial phagocytosis of synapses may contribute to this loss. Stressed neurons can release the nucleotide UTP, which is rapidly converted into UDP, that in turn activates the P2Y6 receptor (P2Y6 R) on the surface of microglia, inducing microglial phagocytosis of neurons. However, whether the activation of P2Y6 R affects microglial phagocytosis of synapses is unknown. We show here that inactivation of P2Y6 R decreases microglial phagocytosis of isolated synapses (synaptosomes) and synaptic loss in neuronal-glial co-cultures. In vivo, wild-type mice aged from 4 to 17 months exhibited reduced synaptic density in cortical and hippocampal regions, which correlated with increased internalization of synaptic material within microglia. However, this aging-induced synaptic loss and internalization were absent in P2Y6 R knockout mice, and these mice also lacked any aging-induced memory loss. Thus, P2Y6 R appears to mediate aging-induced loss of synapses and memory by increasing microglial phagocytosis of synapses. Consequently, blocking P2Y6 R has the potential to prevent age-associated memory impairment.
Collapse
Affiliation(s)
- Jacob M. Dundee
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| | - Mar Puigdellívol
- Department of BiochemistryUniversity of CambridgeCambridgeUK,Institute of NeurosciencesUniversity of BarcelonaBarcelonaSpain
| | - Richard Butler
- The Wellcome Trust Cancer Research UK Gurdon InstituteUniversity of CambridgeCambridgeUK
| | | | - Guy C. Brown
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| |
Collapse
|