1
|
Perdok A, Van Acker ZP, Vrancx C, Sannerud R, Vorsters I, Verrengia A, Callaerts-Végh Z, Creemers E, Gutiérrez Fernández S, D'hauw B, Serneels L, Wierda K, Chávez-Gutiérrez L, Annaert W. Altered expression of Presenilin2 impacts endolysosomal homeostasis and synapse function in Alzheimer's disease-relevant brain circuits. Nat Commun 2024; 15:10412. [PMID: 39613768 DOI: 10.1038/s41467-024-54777-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024] Open
Abstract
Rare mutations in the gene encoding presenilin2 (PSEN2) are known to cause familial Alzheimer's disease (FAD). Here, we explored how altered PSEN2 expression impacts on the amyloidosis, endolysosomal abnormalities, and synaptic dysfunction observed in female APP knock-in mice. We demonstrate that PSEN2 knockout (KO) as well as the FAD-associated N141IKI mutant accelerate AD-related pathologies in female mice. Both models showed significant deficits in working memory that linked to elevated PSEN2 expression in the hippocampal CA3 region. The mossy fiber circuit of APPxPSEN2KO and APPxFADPSEN2 mice had smaller pre-synaptic compartments, distinct changes in synaptic vesicle populations and significantly impaired long term potentiation compared to APPKI mice. At the cellular level, altered PSEN2 expression resulted in endolysosomal defects and lowered surface expression of synaptic proteins. As PSEN2/γ-secretase is restricted to late endosomes/lysosomes, we propose PSEN2 impacts endolysosomal homeostasis, affecting synaptic signaling in AD-relevant vulnerable brain circuits; which could explain how mutant PSEN2 accelerates AD pathogenesis.
Collapse
Affiliation(s)
- Anika Perdok
- Laboratory for Membrane Trafficking, VIB Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Herestraat 49box 602, Leuven, Belgium
| | - Zoë P Van Acker
- Laboratory for Membrane Trafficking, VIB Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Herestraat 49box 602, Leuven, Belgium
| | - Céline Vrancx
- Laboratory for Membrane Trafficking, VIB Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Herestraat 49box 602, Leuven, Belgium
| | - Ragna Sannerud
- Laboratory for Membrane Trafficking, VIB Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Herestraat 49box 602, Leuven, Belgium
| | - Inge Vorsters
- Laboratory for Membrane Trafficking, VIB Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Herestraat 49box 602, Leuven, Belgium
| | - Assunta Verrengia
- Laboratory for Membrane Trafficking, VIB Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Herestraat 49box 602, Leuven, Belgium
| | - Zsuzsanna Callaerts-Végh
- mINT Animal Behavior Facility, Faculty of Psychology, KU Leuven, Tiensestraat 102, Leuven, Belgium
| | - Eline Creemers
- Electrophysiology Expertise Unit, VIB-Center for Brain and Disease Research, Leuven, Belgium
| | - Sara Gutiérrez Fernández
- Department of Neurosciences, KU Leuven, Herestraat 49box 602, Leuven, Belgium
- Laboratory of Proteolytic Mechanisms mediating Neurodegeneration, Leuven, Belgium
| | - Britt D'hauw
- Electrophysiology Expertise Unit, VIB-Center for Brain and Disease Research, Leuven, Belgium
| | - Lutgarde Serneels
- Department of Neurosciences, KU Leuven, Herestraat 49box 602, Leuven, Belgium
- Mouse Expertise Unit, VIB-Center for Brain and Disease Research, Leuven, Belgium
| | - Keimpe Wierda
- Electrophysiology Expertise Unit, VIB-Center for Brain and Disease Research, Leuven, Belgium
| | - Lucía Chávez-Gutiérrez
- Department of Neurosciences, KU Leuven, Herestraat 49box 602, Leuven, Belgium
- Laboratory of Proteolytic Mechanisms mediating Neurodegeneration, Leuven, Belgium
| | - Wim Annaert
- Laboratory for Membrane Trafficking, VIB Center for Brain and Disease Research, Leuven, Belgium.
- Department of Neurosciences, KU Leuven, Herestraat 49box 602, Leuven, Belgium.
| |
Collapse
|
2
|
Jia H, Zhang L, Liao H, Li Y, Liu P, Shi Q, Jiang B, Zhang X, Jiang Y, Nie Z, Jiang M. Association between calculated remnant cholesterol levels and incident risks of Alzheimer's disease among elderly patients with type 2 diabetes: a real-world study. Front Endocrinol (Lausanne) 2024; 15:1505234. [PMID: 39678194 PMCID: PMC11637845 DOI: 10.3389/fendo.2024.1505234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 10/25/2024] [Indexed: 12/17/2024] Open
Abstract
Objective Alzheimer's disease (AD) is a leading cause of dementia, with a rising global burden. Remnant cholesterol (RC), a component of triglyceride-rich lipoproteins, has been implicated in cardiovascular diseases and metabolic disorders, but its role in AD remains unclear. This study investigated the association between RC levels and the risk of AD among elderly patients with type 2 diabetes (T2D) in a real-world clinical setting. Methods We conducted a retrospective cohort study using electronic medical records from Gongli Hospital of Shanghai Pudong New Area, covering the period from 2013 to 2023. The study included 15,364 elderly patients aged 65-80 years with T2D. RC levels were calculated using the equation. The primary outcome was the diagnosis of AD, validated by neurologists using ICD-10-CM code G30. Cox proportional hazards models were employed to estimate hazard ratios (HRs) for AD across quartiles of RC levels, adjusting for potential confounders. Results Over a mean follow-up of 3.69 ± 1.33 years, 312 new cases of AD were identified. A U-shaped relationship was observed between RC levels and AD risk, with the lowest risk associated with RC levels between 0.58-0.64 mmol/L. Both lower (<0.52 mmol/L) and higher (≥0.77 mmol/L) RC levels were linked to increased AD risk. Compared to the reference group (Q2: 0.52-0.64 mmol/L), the adjusted HRs (95% CI) for the lowest and highest quartiles were 1.891 (1.368-2.613) and 1.891 (1.363-2.622), respectively. Each 1 mmol/L increase in RC was associated with a 3.47-fold higher risk of AD (HR=4.474, 95% CI 2.330-8.592). Conclusion RC levels may serve as a predictive biomarker for AD risk, with both extremes posing a higher risk. Future studies should explore the mechanistic pathways and potential interventions targeting RC to prevent AD in high-risk populations.
Collapse
Affiliation(s)
- Huimeng Jia
- Department of General Medicine, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Liuyu Zhang
- Department of General Medicine, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Huijuan Liao
- Department of General Medicine, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Yiming Li
- Department of General Medicine, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Pan Liu
- Department of General Medicine, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Qin Shi
- Department of General Medicine, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Bo Jiang
- Department of General Medicine, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Xian Zhang
- Department of Neurology, Haishu District People’s Hospital, Ningbo, China
| | - Yufeng Jiang
- Department of Cardiology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Zhihong Nie
- Department of General Medicine, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Mei Jiang
- Department of Neurology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| |
Collapse
|
3
|
Hao YA, Lee S, Roth RH, Natale S, Gomez L, Taxidis J, O'Neill PS, Villette V, Bradley J, Wang Z, Jiang D, Zhang G, Sheng M, Lu D, Boyden E, Delvendahl I, Golshani P, Wernig M, Feldman DE, Ji N, Ding J, Südhof TC, Clandinin TR, Lin MZ. A fast and responsive voltage indicator with enhanced sensitivity for unitary synaptic events. Neuron 2024; 112:3680-3696.e8. [PMID: 39305894 PMCID: PMC11581914 DOI: 10.1016/j.neuron.2024.08.019] [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: 10/10/2023] [Revised: 07/23/2024] [Accepted: 08/27/2024] [Indexed: 09/29/2024]
Abstract
A remaining challenge for genetically encoded voltage indicators (GEVIs) is the reliable detection of excitatory postsynaptic potentials (EPSPs). Here, we developed ASAP5 as a GEVI with enhanced activation kinetics and responsivity near resting membrane potentials for improved detection of both spiking and subthreshold activity. ASAP5 reported action potentials (APs) in vivo with higher signal-to-noise ratios than previous GEVIs and successfully detected graded and subthreshold responses to sensory stimuli in single two-photon trials. In cultured rat or human neurons, somatic ASAP5 reported synaptic events propagating centripetally and could detect ∼1-mV EPSPs. By imaging spontaneous EPSPs throughout dendrites, we found that EPSP amplitudes decay exponentially during propagation and that amplitude at the initiation site generally increases with distance from the soma. These results extend the applications of voltage imaging to the quantal response domain, including in human neurons, opening up the possibility of high-throughput, high-content characterization of neuronal dysfunction in disease.
Collapse
Affiliation(s)
- Yukun A Hao
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Neurobiology, Stanford University, Stanford, CA 94305, USA
| | - Sungmoo Lee
- Department of Neurobiology, Stanford University, Stanford, CA 94305, USA
| | - Richard H Roth
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Silvia Natale
- Department of Molecular & Cellular Physiology, Stanford University, Stanford, CA 94305, USA
| | - Laura Gomez
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA; Department of Physics, University of California Berkeley, CA 94720, USA
| | - Jiannis Taxidis
- Department of Neurology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Philipp S O'Neill
- Department of Molecular Life Sciences, University of Zurich (UZH), 8057 Zurich, Switzerland; Neuroscience Center Zurich, 8057 Zurich, Switzerland
| | - Vincent Villette
- Institut de Biologie de l'École Normale Supérieure (IBENS), CNRS, INSERM, PSL Research University, Paris 75005, France
| | - Jonathan Bradley
- Institut de Biologie de l'École Normale Supérieure (IBENS), CNRS, INSERM, PSL Research University, Paris 75005, France
| | - Zeguan Wang
- Departments of Brain and Cognitive Sciences, Media Arts and Sciences, and Biological Engineering, MIT, Cambridge, MA 02139, USA; McGovern Institute, MIT, Cambridge, MA 02139, USA
| | - Dongyun Jiang
- Department of Neurobiology, Stanford University, Stanford, CA 94305, USA
| | - Guofeng Zhang
- Department of Neurobiology, Stanford University, Stanford, CA 94305, USA
| | - Mengjun Sheng
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Di Lu
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Edward Boyden
- Departments of Brain and Cognitive Sciences, Media Arts and Sciences, and Biological Engineering, MIT, Cambridge, MA 02139, USA; McGovern Institute, MIT, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Cambridge, MA 02139, USA
| | - Igor Delvendahl
- Department of Molecular Life Sciences, University of Zurich (UZH), 8057 Zurich, Switzerland; Neuroscience Center Zurich, 8057 Zurich, Switzerland
| | - Peyman Golshani
- Department of Neurology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA; Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Marius Wernig
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Daniel E Feldman
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Na Ji
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA; Department of Physics, University of California Berkeley, CA 94720, USA
| | - Jun Ding
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Thomas C Südhof
- Department of Molecular & Cellular Physiology, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Thomas R Clandinin
- Department of Neurobiology, Stanford University, Stanford, CA 94305, USA
| | - Michael Z Lin
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Neurobiology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
4
|
Liu X, Chen P, Wu W, Zhong M, Dong S, Lin H, Dai C, Zhang Z, Lin S, Che C, Xu J, Li C, Li H, Pan X, Chen Z, Chen X, Ye ZC. Compound (E)-2-(3,4-dihydroxystyryl)-3-hydroxy-4H-pyran-4-one downregulation of Galectin-3 ameliorates Aβ pathogenesis-induced neuroinflammation in 5 × FAD mice. Life Sci 2024; 357:123085. [PMID: 39362584 DOI: 10.1016/j.lfs.2024.123085] [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: 07/11/2024] [Revised: 09/02/2024] [Accepted: 09/28/2024] [Indexed: 10/05/2024]
Abstract
AIMS Alzheimer's disease (AD) is characterized by β-amyloid (Aβ) aggregation and neuroinflammation, leading to progressive synaptic loss and cognitive decline. Recent evidence suggests that Galectin-3 (Gal-3) plays a critical role in Aβ pathogenesis. However, strategies to simultaneously target Gal-3 and Aβ are currently insufficient. This study evaluates the therapeutic efficacy of (E)-2-(3,4-dihydroxystyryl)-3-hydroxy-4H-pyran-4-one (D30), in reducing Gal-3 and Aβ pathogenesis. MATERIALS AND METHODS We applied exogenous oligomeric Aβ and used 5 × FAD mice to assess the impact of Aβ on Gal-3 deposition, microglial activation, and cognitive function. Thy1-EGFP mice were employed to observe dendritic spines. Comprehensive evaluations of D30's effects included behavioral studies, transcriptomic analysis, Western blotting, and immunofluorescent staining. The interaction between D30 and Gal-3 was examined using fluorescence resonance energy transfer (FRET) and microscale thermophoresis (MST). KEY FINDINGS D30 effectively reduced Aβ monomer production by inhibiting Amyloid Precursor Protein (APP) and presenilin 1 (PS1) expression, and decreased Aβ aggregation. Treatment with D30 improved cognitive functions, reversed dendritic spine loss, and increased PSD95 expression in 5 × FAD mice. Additionally, D30 significantly lowered Gal-3 levels in both plasma and hippocampal tissues. D30 binds to Gal-3 and disrupts the interaction between Gal-3 and TREM2, as confirmed by FRET and MST. SIGNIFICANCE Our findings underscore the interaction between Gal-3 and Aβ in AD and its role in systemic inflammation using the 5 × FAD mouse model. Being able to target and regulate Gal-3 together with Aβ is crucial for preventing neuroinflammation and protecting synapses, D30 emerged as a novel compound with promising potential for AD treatment.
Collapse
Affiliation(s)
- Xueyan Liu
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China; School of Pharmacy, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Ping Chen
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China; Department of Anesthesiology, Anesthesiology Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian Province, China
| | - Wei Wu
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Meihua Zhong
- School of Pharmacy, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Shiyu Dong
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Huiling Lin
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Chaoxian Dai
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Zhile Zhang
- School of Pharmacy, Fujian Medical University, Fuzhou 350112, Fujian Province, China; Ningde Rehabilitation Hospital, Ningde 352105, Fujian Province, China
| | - Shiqi Lin
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Cuilan Che
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Jiexin Xu
- School of Pharmacy, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Chenlu Li
- Department of Hyperbaric Oxygen, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, Fujian Province, China
| | - Hongwei Li
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun 130012, Jilin Province, China
| | - Xiaodong Pan
- Department of Neurology, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University, Fujian Province, China
| | - Zhou Chen
- Overseas Education College of Fujian Medical University, Fujian Medical University, Fuzhou 350004, Fujian Province, China
| | - Xiaochun Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University, Fujian Province, China.
| | - Zu-Cheng Ye
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China.
| |
Collapse
|
5
|
Paiva I, Seguin J, Grgurina I, Singh AK, Cosquer B, Plassard D, Tzeplaeff L, Le Gras S, Cotellessa L, Decraene C, Gambi J, Alcala-Vida R, Eswaramoorthy M, Buée L, Cassel JC, Giacobini P, Blum D, Merienne K, Kundu TK, Boutillier AL. Dysregulated expression of cholesterol biosynthetic genes in Alzheimer's disease alters epigenomic signatures of hippocampal neurons. Neurobiol Dis 2024; 198:106538. [PMID: 38789057 DOI: 10.1016/j.nbd.2024.106538] [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: 12/29/2023] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 05/26/2024] Open
Abstract
Aging is the main risk factor of cognitive neurodegenerative diseases such as Alzheimer's disease, with epigenome alterations as a contributing factor. Here, we compared transcriptomic/epigenomic changes in the hippocampus, modified by aging and by tauopathy, an AD-related feature. We show that the cholesterol biosynthesis pathway is severely impaired in hippocampal neurons of tauopathic but not of aged mice pointing to vulnerability of these neurons in the disease. At the epigenomic level, histone hyperacetylation was observed at neuronal enhancers associated with glutamatergic regulations only in the tauopathy. Lastly, a treatment of tau mice with the CSP-TTK21 epi-drug that restored expression of key cholesterol biosynthesis genes counteracted hyperacetylation at neuronal enhancers and restored object memory. As acetyl-CoA is the primary substrate of both pathways, these data suggest that the rate of the cholesterol biosynthesis in hippocampal neurons may trigger epigenetic-driven changes, that may compromise the functions of hippocampal neurons in pathological conditions.
Collapse
Affiliation(s)
- Isabel Paiva
- University of Strasbourg, Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France; CNRS, UMR7364 - Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France.
| | - Jonathan Seguin
- University of Strasbourg, Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France; CNRS, UMR7364 - Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France
| | - Iris Grgurina
- University of Strasbourg, Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France; CNRS, UMR7364 - Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France
| | - Akash Kumar Singh
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, India
| | - Brigitte Cosquer
- University of Strasbourg, Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France; CNRS, UMR7364 - Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France
| | - Damien Plassard
- University of Strasbourg, CNRS UMR7104, Inserm U1258 - GenomEast Platform - IGBMC - Institut de Génétique et de Biologie Moléculaire et Cellulaire, F-67404 Illkirch, France
| | - Laura Tzeplaeff
- University of Strasbourg, Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France; CNRS, UMR7364 - Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France
| | - Stephanie Le Gras
- University of Strasbourg, CNRS UMR7104, Inserm U1258 - GenomEast Platform - IGBMC - Institut de Génétique et de Biologie Moléculaire et Cellulaire, F-67404 Illkirch, France
| | - Ludovica Cotellessa
- University of Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, FHU 1000 Days for Health, 59000 Lille, France
| | - Charles Decraene
- University of Strasbourg, Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France; CNRS, UMR7364 - Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France
| | - Johanne Gambi
- University of Strasbourg, Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France; CNRS, UMR7364 - Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France
| | - Rafael Alcala-Vida
- University of Strasbourg, Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France; CNRS, UMR7364 - Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France
| | - Muthusamy Eswaramoorthy
- Chemistry and Physics of Materials Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Luc Buée
- University of Lille, Inserm, CHU Lille, UMR-S1172 LilNCog - Lille Neuroscience & Cognition, Lille, France; Alzheimer and Tauopathies, LabEx DISTALZ, France
| | - Jean-Christophe Cassel
- University of Strasbourg, Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France; CNRS, UMR7364 - Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France
| | - Paolo Giacobini
- University of Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, FHU 1000 Days for Health, 59000 Lille, France
| | - David Blum
- University of Lille, Inserm, CHU Lille, UMR-S1172 LilNCog - Lille Neuroscience & Cognition, Lille, France; Alzheimer and Tauopathies, LabEx DISTALZ, France
| | - Karine Merienne
- University of Strasbourg, Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France; CNRS, UMR7364 - Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France
| | - Tapas K Kundu
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, India
| | - Anne-Laurence Boutillier
- University of Strasbourg, Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France; CNRS, UMR7364 - Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France.
| |
Collapse
|
6
|
Gao C, Shang J, Sun Z, Xia M, Gao D, Sun R, Li W, Wang F, Zhang J. Presenilin2 D439A Mutation Induces Dysfunction of Mitochondrial Fusion/Fission Dynamics and Abnormal Regulation of GTPase Activity. Mol Neurobiol 2024; 61:5047-5070. [PMID: 38159198 PMCID: PMC11249618 DOI: 10.1007/s12035-023-03858-y] [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: 05/17/2023] [Accepted: 12/04/2023] [Indexed: 01/03/2024]
Abstract
Alzheimer's disease (AD) is an age-related progressive neurodegenerative disease, and approximately 10% of AD cases are early-onset familial AD (EOFAD), which is mainly linked to point mutations in genes encoding presenilins (PS1 and PS2). Mutations in PS2 are extremely rare and have not received enough attention. Recently, studies have found that Rho GTPase activity is closely related to the pathogenesis of AD. In this study, we used transcriptome sequencing in PS2 siRNA-transfected SH-SY5Y cells and found a group of differentially expressed genes (DEGs) related to the regulation of GTPase activity. Among those DEGs, the most significantly downregulated was Rho guanine nucleotide exchange factor 5 (ARHGEF5). GTPase activity in PS2 siRNA-transfected cells was significantly decreased. Then, we found that the expression of ARHGEF5 and the GTPase activity of Mitochondrial Rho GTPase 2 (Miro2) in PS2 D439A mutant SH-SY5Y cells were significantly decreased. We found for the first time that PS2 can bind to Miro2, and the PS2 D439A mutation reduced the binding between PS2 and Miro2, reduced the expression of Miro2, and resulted in an imbalance in mitochondrial fusion/fission dynamics. In conclusion, PS2 gene knockdown may participate in the pathogenesis of AD through the regulation of GTPase activity. The imbalance in mitochondrial dynamics mediated by the PS2 D439A mutation through regulation of the expression and GTPase activity of Miro2 may be a potential pathogenic mechanism of AD.
Collapse
Affiliation(s)
- Chenhao Gao
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Junkui Shang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China
- Department of Neurology, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China
| | - Zhengyu Sun
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China
- Department of Neurology, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China
| | - Mingrong Xia
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China
| | - Dandan Gao
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China
| | - Ruihua Sun
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Wei Li
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China
| | - Fengyu Wang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China
- Department of Neurology, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China
| | - Jiewen Zhang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China.
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450003, Henan, China.
- Department of Neurology, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China.
| |
Collapse
|
7
|
Uchiumi O, Zou J, Yamaki S, Hori Y, Ono M, Yamamoto R, Kato N. Disruption of sphingomyelin synthase 2 gene alleviates cognitive impairment in a mouse model of Alzheimer's disease. Brain Res 2024; 1835:148934. [PMID: 38609029 DOI: 10.1016/j.brainres.2024.148934] [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: 02/15/2024] [Revised: 03/28/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
The membrane raft accommodates the key enzymes synthesizing amyloid β (Aβ). One of the two characteristic components of the membrane raft, cholesterol, is well known to promote the key enzymes that produce amyloid-β (Aβ) and exacerbate Alzheimer's disease (AD) pathogenesis. Given that the raft is a physicochemical platform for the sound functioning of embedded bioactive proteins, the other major lipid component sphingomyelin may also be involved in AD. Here we knocked out the sphingomyelin synthase 2 gene (SMS2) in 3xTg AD model mice by hybridization, yielding SMS2KO mice (4S mice). The novel object recognition test in 9/10-month-old 4S mice showed that cognitive impairment in 3xTg mice was alleviated by SMS2KO, though performance in the Morris water maze (MWM) was not improved. The tail suspension test detected a depressive trait in 4S mice, which may have hindered the manifestation of performance in the wet, stressful environment of MWM. In the hippocampal CA1, hyperexcitability in 3xTg was also found alleviated by SMS2KO. In the hippocampal dentate gyrus of 4S mice, the number of neurons positive with intracellular Aβ or its precursor proteins, the hallmark of young 3xTg mice, is reduced to one-third, suggesting an SMS2KO-led suppression of syntheses of those peptides in the dentate gyrus. Although we previously reported that large-conductance calcium-activated potassium (BK) channels are suppressed in 3xTg mice and their recovery relates to cognitive amelioration, no changes occurred by hybridization. Sphingomyelin in the membrane raft may serve as a novel target for AD drugs.
Collapse
Affiliation(s)
- Osamu Uchiumi
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Jingyu Zou
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan; First Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Sachiko Yamaki
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Yoshie Hori
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Munenori Ono
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Ryo Yamamoto
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Nobuo Kato
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan.
| |
Collapse
|
8
|
Song Z, He J, Yu W, He C, Yang M, Li P, Li Z, Jian G, Cheng S. Exploring the multifaceted therapeutic mechanism of Schisanlactone E (XTS) in APP/PS1 mouse model of Alzheimer's disease through multi-omics analysis. Front Microbiol 2024; 15:1440564. [PMID: 39044957 PMCID: PMC11263214 DOI: 10.3389/fmicb.2024.1440564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 06/28/2024] [Indexed: 07/25/2024] Open
Abstract
Background Schisanlactone E, also known as XueTongSu (XTS), is an active compound extracted from the traditional Tujia medicine Kadsura heteroclita ("XueTong"). Recent studies highlight its anti-inflammatory and antioxidant properties, yet the mechanisms of XTS's therapeutic effects on Alzheimer's disease (AD) are unclear. This study aims to elucidate the therapeutic efficacy and mechanisms of XTS in AD. Methods Ten C57BL/6 mice were assigned to the control group (NC), and twenty APP/PS1 transgenic mice were randomly divided into the model group (M) (10 mice) and the XTS treatment group (Tre) (10 mice). After an acclimatization period of 7 days, intraperitoneal injections were administered over a 60-day treatment period. The NC and M groups received saline, while the Tre group received XTS at 2 mg/kg. Learning and memory abilities were assessed using the Morris Water Maze (MWM) test. Histopathological changes were evaluated using hematoxylin and eosin (HE) and Nissl staining, and immunofluorescence was used to assess pathological products and glial cell activation. Cytokine levels (IL-1β, IL-6, TNF-α) in the hippocampus were quantified by qPCR. 16S rDNA sequencing analyzed gut microbiota metabolic alterations, and metabolomic analysis was performed on cortical samples. The KEGG database was used to analyze the regulatory mechanisms of XTS in AD treatment. Results XTS significantly improved learning and spatial memory in APP/PS1 mice and ameliorated histopathological changes, reducing Aβ plaque aggregation and glial cell activation. XTS decreased the expression of inflammatory cytokines IL-1β, IL-6, and TNF-α. It also enhanced gut microbiota diversity, notably increasing Akkermansia species, and modulated levels of metabolites such as isosakuranetin, 5-KETE, 4-methylcatechol, and sphinganine. Pathway analysis indicated that XTS regulated carbohydrate metabolism, neuroactive ligand-receptor interactions, and alanine, aspartate, and glutamate metabolism, mitigating gut microbiota dysbiosis and metabolic disturbances. Conclusion XTS ameliorates cognitive deficits, pathological changes, and inflammatory responses in APP/PS1 mice. It significantly modulates the gut microbiota, particularly increasing Akkermansia abundance, and influences levels of key metabolites in both the gut and brain. These findings suggest that XTS exerts anti-AD effects through the microbial-gut-brain axis (MGBA).
Collapse
Affiliation(s)
- Zhenyan Song
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jiawei He
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Wenjing Yu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chunxiang He
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Miao Yang
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ping Li
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ze Li
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Gonghui Jian
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Shaowu Cheng
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan University of Chinese Medicine, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
9
|
Bretou M, Sannerud R, Escamilla-Ayala A, Leroy T, Vrancx C, Van Acker ZP, Perdok A, Vermeire W, Vorsters I, Van Keymolen S, Maxson M, Pavie B, Wierda K, Eskelinen EL, Annaert W. Accumulation of APP C-terminal fragments causes endolysosomal dysfunction through the dysregulation of late endosome to lysosome-ER contact sites. Dev Cell 2024; 59:1571-1592.e9. [PMID: 38626765 DOI: 10.1016/j.devcel.2024.03.030] [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: 10/18/2021] [Revised: 11/02/2023] [Accepted: 03/20/2024] [Indexed: 04/18/2024]
Abstract
Neuronal endosomal and lysosomal abnormalities are among the early changes observed in Alzheimer's disease (AD) before plaques appear. However, it is unclear whether distinct endolysosomal defects are temporally organized and how altered γ-secretase function or amyloid precursor protein (APP) metabolism contribute to these changes. Inhibiting γ-secretase chronically, in mouse embryonic fibroblast and hippocampal neurons, led to a gradual endolysosomal collapse initiated by decreased lysosomal calcium and increased cholesterol, causing downstream defects in endosomal recycling and maturation. This endolysosomal demise is γ-secretase dependent, requires membrane-tethered APP cytoplasmic domains, and is rescued by APP depletion. APP C-terminal fragments (CTFs) localized to late endosome/lysosome-endoplasmic reticulum contacts; an excess of APP-CTFs herein reduced lysosomal Ca2+ refilling from the endoplasmic reticulum, promoting cholesterol accretion. Tonic regulation by APP-CTFs provides a mechanistic explanation for their cellular toxicity: failure to timely degrade APP-CTFs sustains downstream signaling, instigating lysosomal dyshomeostasis, as observed in prodromal AD. This is the opposite of substrates such as Notch, which require intramembrane proteolysis to initiate signaling.
Collapse
Affiliation(s)
- Marine Bretou
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Ragna Sannerud
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | | | - Tom Leroy
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Céline Vrancx
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Zoë P Van Acker
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Anika Perdok
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Wendy Vermeire
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Inge Vorsters
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Sophie Van Keymolen
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Michelle Maxson
- Cell Biology Program, The Hospital for Sick Children, Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Benjamin Pavie
- VIB-BioImaging Core, VIB-Center for Brain and Disease Research, Leuven, Belgium
| | - Keimpe Wierda
- Electrophysiology Expertise Unit, VIB-Center for Brain and Disease Research, Leuven, Belgium
| | | | - Wim Annaert
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
10
|
Deng Y, Liang X, Li Y, Jiang L, Wang J, Tang J, Li J, Xie Y, Xiao K, Zhu P, Guo Y, Luo Y, Tang Y. PGC-1α in the hippocampus mediates depressive-like and stress-coping behaviours and regulates excitatory synapses in the dentate gyrus in mice. Neuropharmacology 2024; 250:109908. [PMID: 38492883 DOI: 10.1016/j.neuropharm.2024.109908] [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: 09/17/2023] [Revised: 02/13/2024] [Accepted: 03/10/2024] [Indexed: 03/18/2024]
Abstract
Decreased hippocampal synaptic plasticity is an important pathological change in stress-related mood disorders, including major depressive disorder. However, the underlying mechanism is unclear. PGC-1α, a transcriptional coactivator, is a key factor in synaptic plasticity. We investigated the relationships between changes in hippocampal PGC-1α expression and depressive-like and stress-coping behaviours, and whether they are related to hippocampal synapses. Adeno-associated virus was used to alter hippocampal PGC-1α expression in male C57BL/6 mice. The sucrose preference test and forced swimming test were used to assess their depressive-like and stress-coping behaviours, respectively. Immunohistochemistry and stereology were used to calculate the total number of excitatory synapses in each hippocampal subregion (the cornu ammonis (CA) 1, CA3, and dentate gyrus). Immunofluorescence was used to visualize the changes in dendritic structure. Western blotting was used to detect the expression of hippocampal PGC-1α and mitochondrial-associated proteins, such as UCP2, NRF1 and mtTFAs. Our results showed that mice with downregulated PGC-1α expression in the hippocampus exhibited depressive-like and passive stress-coping behaviours, while mice with upregulated PGC-1α in the hippocampus exhibited increased stress-coping behaviours. Moreover, the downregulation of hippocampal PGC-1α expression resulted in a decrease in the number of excitatory synapses in the DG and in the protein expression of UCP2 in the hippocampus. Alternatively, upregulation of hippocampal PGC-1α yielded the opposite results. This suggests that hippocampal PGC-1α is involved in regulating depressive-like and stress-coping behaviours and modulating the number of excitatory synapses in the DG. This provides new insight for the development of antidepressants.
Collapse
Affiliation(s)
- Yuhui Deng
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Xin Liang
- Department of Pathology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yue Li
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Lin Jiang
- Lab Teaching and Management Center, Chongqing Medical University, Chongqing, 400016, PR China
| | - Jin Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Jing Tang
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Jing Li
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yuhan Xie
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Kai Xiao
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Peilin Zhu
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yijing Guo
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yanmin Luo
- Department of Physiology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China.
| | - Yong Tang
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China.
| |
Collapse
|
11
|
Gong Y, Li M, Liu M, Wu X, Li Y, Qin C, Zhang L. Apolipoprotein E4 interferes with lipid metabolism to exacerbate depression-like behaviors in 5xFAD mice. Animal Model Exp Med 2024; 7:347-361. [PMID: 38895818 PMCID: PMC11228103 DOI: 10.1002/ame2.12446] [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: 05/07/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Apolipoprotein E4 (ApoE4) allele is the strongest genetic risk factor for late-onset Alzheimer's disease, and it can aggravate depressive symptoms in non-AD patients. However, the impact of ApoE4 on AD-associated depression-like behaviors and its underlying pathogenic mechanisms remain unclear. METHODS This study developed a 5xFAD mouse model overexpressing human ApoE4 (E4FAD). Behavioral assessments and synaptic function tests were conducted to explore the effects of ApoE4 on cognition and depression in 5xFAD mice. Changes in peripheral and central lipid metabolism, as well as the levels of serotonin (5-HT) and γ-aminobutyric acid (GABA) neurotransmitters in the prefrontal cortex, were examined. In addition, the protein levels of 24-dehydrocholesterol reductase/glycogen synthase kinase-3 beta/mammalian target of rapamycin (DHCR24/GSK3β/mTOR) and postsynaptic density protein 95/calmodulin-dependent protein kinase II/brain-derived neurotrophic factor (PSD95/CaMK-II/BDNF) were measured to investigate the molecular mechanism underlying the effects of ApoE4 on AD mice. RESULTS Compared with 5xFAD mice, E4FAD mice exhibited more severe depression-like behaviors and cognitive impairments. These mice also exhibited increased amyloid-beta deposition in the hippocampus, increased astrocyte numbers, and decreased expression of depression-related neurotransmitters 5-HT and GABA in the prefrontal cortex. Furthermore, lipid metabolism disorders were observed in E4FAD, manifesting as elevated low-density lipoprotein cholesterol and reduced high-density lipoprotein cholesterol in peripheral blood, decreased cholesterol level in the prefrontal cortex, and reduced expression of key enzymes and proteins related to cholesterol synthesis and homeostasis. Abnormal expression of proteins related to the DHCR24/GSK3β/mTOR and PSD95/CaMK-II/BDNF pathways was also observed. CONCLUSION This study found that ApoE4 overexpression exacerbates depression-like behaviors in 5xFAD mice and confirmed that ApoE4 reduces cognitive function in these mice. The mechanism may involve the induction of central and peripheral lipid metabolism disorders. Therefore, modulating ApoE expression or function to restore cellular lipid homeostasis may be a promising therapeutic target for AD comorbid with depression. This study also provided a better animal model for studying AD comorbid with depression.
Collapse
Affiliation(s)
- Yanju Gong
- iNHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and lnnovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PuMC)BeijingChina
| | - Mingfeng Li
- iNHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and lnnovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PuMC)BeijingChina
| | - Min Liu
- iNHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and lnnovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PuMC)BeijingChina
| | - Xinghan Wu
- iNHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and lnnovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PuMC)BeijingChina
| | - Yanhong Li
- iNHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and lnnovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PuMC)BeijingChina
| | - Chuan Qin
- iNHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and lnnovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PuMC)BeijingChina
- Changping National Laboratory (CPNL)BeijingChina
| | - Ling Zhang
- iNHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and lnnovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PuMC)BeijingChina
| |
Collapse
|
12
|
Sengupta S, Yaeger JD, Schultz MM, Francis KR. Dishevelled localization and function are differentially regulated by structurally distinct sterols. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.593701. [PMID: 38798572 PMCID: PMC11118412 DOI: 10.1101/2024.05.14.593701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The Dishevelled (DVL) family of proteins form supramolecular protein and lipid complexes at the cytoplasmic interface of the plasma membrane to regulate tissue patterning, proliferation, cell polarity, and oncogenic processes through DVL-dependent signaling, such as Wnt/β-catenin. While DVL binding to cholesterol is required for its membrane association, the specific structural requirements and cellular impacts of DVL-sterol association are unclear. We report that intracellular sterols which accumulate within normal and pathological conditions cause aberrant DVL activity. In silico and molecular analyses suggested orientation of the β- and α-sterol face within the DVL-PDZ domain regulates DVL-sterol binding. Intracellular accumulation of naturally occurring sterols impaired DVL2 plasma membrane association, inducing DVL2 nuclear localization via Foxk2. Changes to intracellular sterols also selectively impaired DVL2 protein-protein interactions This work identifies sterol specificity as a regulator of DVL signaling, suggests intracellular sterols cause distinct impacts on DVL activity, and supports a role for intracellular sterol homeostasis in cell signaling.
Collapse
Affiliation(s)
- Sonali Sengupta
- Cellular Therapies and Stem Cell Biology Group, Sanford Research, Sioux Falls, SD, 57104, USA
| | - Jazmine D.W. Yaeger
- Cellular Therapies and Stem Cell Biology Group, Sanford Research, Sioux Falls, SD, 57104, USA
| | - Maycie M. Schultz
- Cellular Therapies and Stem Cell Biology Group, Sanford Research, Sioux Falls, SD, 57104, USA
| | - Kevin R. Francis
- Cellular Therapies and Stem Cell Biology Group, Sanford Research, Sioux Falls, SD, 57104, USA
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, SD, 57105, USA
| |
Collapse
|
13
|
Area-Gomez E, Schon EA. Towards a Unitary Hypothesis of Alzheimer's Disease Pathogenesis. J Alzheimers Dis 2024; 98:1243-1275. [PMID: 38578892 DOI: 10.3233/jad-231318] [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] [Indexed: 04/07/2024]
Abstract
The "amyloid cascade" hypothesis of Alzheimer's disease (AD) pathogenesis invokes the accumulation in the brain of plaques (containing the amyloid-β protein precursor [AβPP] cleavage product amyloid-β [Aβ]) and tangles (containing hyperphosphorylated tau) as drivers of pathogenesis. However, the poor track record of clinical trials based on this hypothesis suggests that the accumulation of these peptides is not the only cause of AD. Here, an alternative hypothesis is proposed in which the AβPP cleavage product C99, not Aβ, is the main culprit, via its role as a regulator of cholesterol metabolism. C99, which is a cholesterol sensor, promotes the formation of mitochondria-associated endoplasmic reticulum (ER) membranes (MAM), a cholesterol-rich lipid raft-like subdomain of the ER that communicates, both physically and biochemically, with mitochondria. We propose that in early-onset AD (EOAD), MAM-localized C99 is elevated above normal levels, resulting in increased transport of cholesterol from the plasma membrane to membranes of intracellular organelles, such as ER/endosomes, thereby upregulating MAM function and driving pathology. By the same token, late-onset AD (LOAD) is triggered by any genetic variant that increases the accumulation of intracellular cholesterol that, in turn, boosts the levels of C99 and again upregulates MAM function. Thus, the functional cause of AD is upregulated MAM function that, in turn, causes the hallmark disease phenotypes, including the plaques and tangles. Accordingly, the MAM hypothesis invokes two key interrelated elements, C99 and cholesterol, that converge at the MAM to drive AD pathogenesis. From this perspective, AD is, at bottom, a lipid disorder.
Collapse
Affiliation(s)
- Estela Area-Gomez
- Department of Neurology, Columbia University, New York, NY, USA
- Centro de Investigaciones Biológicas "Margarita Salas", Spanish National Research Council, Madrid, Spain
| | - Eric A Schon
- Department of Neurology, Columbia University, New York, NY, USA
- Department of Genetics and Development>, Columbia University, New York, NY, USA
| |
Collapse
|
14
|
Frankel EB, Tiroumalechetty A, Henry PS, Su Z, Wu Y, Kurshan PT. Protein-lipid interactions drive presynaptic assembly upstream of cell adhesion molecules. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.17.567618. [PMID: 38014115 PMCID: PMC10680821 DOI: 10.1101/2023.11.17.567618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Textbook models of synaptogenesis position cell adhesion molecules such as neurexin as initiators of synapse assembly. Here we discover a mechanism for presynaptic assembly that occurs prior to neurexin recruitment, while supporting a role for neurexin in synapse maintenance. We find that the cytosolic active zone scaffold SYD-1 interacts with membrane phospholipids to promote active zone protein clustering at the plasma membrane, and subsequently recruits neurexin to stabilize those clusters. Employing molecular dynamics simulations to model intrinsic interactions between SYD-1 and lipid bilayers followed by in vivo tests of these predictions, we find that PIP2-interacting residues in SYD-1's C2 and PDZ domains are redundantly necessary for proper active zone assembly. Finally, we propose that the uncharacterized yet evolutionarily conserved short γ isoform of neurexin represents a minimal neurexin sequence that can stabilize previously assembled presynaptic clusters, potentially a core function of this critical protein.
Collapse
Affiliation(s)
- Elisa B Frankel
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461
| | | | - Parise S Henry
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Zhaoqian Su
- Data Science Institute, Vanderbilt University, 1001 19th Ave S, Nashville, TN, 37212
| | - Yinghao Wu
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Peri T Kurshan
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461
- Lead Contact
| |
Collapse
|
15
|
Fu WY, Fu AKY, Ip NY. Neuronal γ-secretase regulates synaptic functions via cholesterol homeostasis. Neuron 2023; 111:3133-3135. [PMID: 37857086 DOI: 10.1016/j.neuron.2023.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023]
Abstract
In this issue of Neuron, Essayan-Perez and Südhof1 demonstrate roles for γ-secretase in the regulation of synaptic functions in human neurons. Chronic attenuation of γ-secretase activity increases synapse formation but decreases neurotransmission (i.e., the probability of presynaptic release), likely due to impairment of cholesterol metabolism.
Collapse
Affiliation(s)
- Wing-Yu Fu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
| | - Amy K Y Fu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China; Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen 518057, Guangdong, China
| | - Nancy Y Ip
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China; Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen 518057, Guangdong, China.
| |
Collapse
|
16
|
Zhao X, Zhang S, Sanders AR, Duan J. Brain Lipids and Lipid Droplet Dysregulation in Alzheimer's Disease and Neuropsychiatric Disorders. Complex Psychiatry 2023; 9:154-171. [PMID: 38058955 PMCID: PMC10697751 DOI: 10.1159/000535131] [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: 07/11/2023] [Accepted: 11/06/2023] [Indexed: 12/08/2023] Open
Abstract
Background Lipids are essential components of the structure and for the function of brain cells. The intricate balance of lipids, including phospholipids, glycolipids, cholesterol, cholesterol ester, and triglycerides, is crucial for maintaining normal brain function. The roles of lipids and lipid droplets and their relevance to neurodegenerative and neuropsychiatric disorders (NPDs) remain largely unknown. Summary Here, we reviewed the basic role of lipid components as well as a specific lipid organelle, lipid droplets, in brain function, highlighting the potential impact of altered lipid metabolism in the pathogenesis of Alzheimer's disease (AD) and NDPs. Key Messages Brain lipid dysregulation plays a pivotal role in the pathogenesis and progression of neurodegenerative and NPDs including AD and schizophrenia. Understanding the cell type-specific mechanisms of lipid dysregulation in these diseases is crucial for identifying better diagnostic biomarkers and for developing therapeutic strategies aiming at restoring lipid homeostasis.
Collapse
Affiliation(s)
- Xiaojie Zhao
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, USA
| | - Siwei Zhang
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, USA
| | - Alan R. Sanders
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, USA
| | - Jubao Duan
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, USA
| |
Collapse
|