1
|
Saleh NK, Farrag SM, El-Yamany MF, Kamel AS. Exploring Dapagliflozin's Influence on Autophagic Flux in Mania-like Behaviour: Insights from the LKB1/AMPK/LC3 Pathway in a Mouse Model. J Neuroimmune Pharmacol 2025; 20:57. [PMID: 40402300 PMCID: PMC12098488 DOI: 10.1007/s11481-025-10218-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 04/30/2025] [Indexed: 05/23/2025]
Abstract
Mania-like episodes are neuropsychiatric disturbances associated with bipolar disorder (BD). Autophagic flux disturbance evolved as one of the molecular mechanisms implicated in mania. Recently, Dapagliflozin (DAPA) has corrected autophagic signaling in several neurological disorders. Yet, no endeavours examined the autophagic impact of DAPA in mania-like behaviours. This study aimed to investigate the effect of DAPA on disrupted autophagic pathways in a mouse model of mania-like behaviour. Mania-like behaviour was induced through paradoxical sleep deprivation (PSD) using the multiple-platform method for a duration of 36 h. Mice were divided into three groups, with DAPA (1 mg/kg/day, orally) administered for one week. Behavioural assessments were conducted on the 7th day. DAPA mitigated anxiety-like behaviour in the open field test and improved motor coordination and muscle tone in the rotarod test. Mechanistically, DAPA activated hippocampal autophagy-related markers; liver kinase B1/AMP-activated protein kinase (LKB1/AMPK) pathway, autophagy related gene 7 (ATG7), and microtubule-associated protein light chain 3II (LC3II). This was associated with reduced levels of the autophagosome receptor p62 protein, which subsequently enhanced GABAA receptor-associated protein (GABARAP), facilitating the surface presentation of GABAA receptors. Additionally, DAPA upregulated the GABAB receptor R2 subunit through trophic factors such as brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF). Furthermore, DAPA mitigated elevated serum stress hormones and restored the balance between proinflammatory and anti-inflammatory cytokines in both cortical and hippocampal tissues. These findings highlight the role of autophagic flux modulation by DAPA and its therapeutic potential in mitigating mania-like behaviours.
Collapse
Affiliation(s)
- Nada K Saleh
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Sama M Farrag
- Pharmacology and Toxicology Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST), Giza, Egypt
| | - Mohamed F El-Yamany
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Ahmed S Kamel
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| |
Collapse
|
2
|
Yang L, Guo C, Zheng Z, Dong Y, Xie Q, Lv Z, Li M, Lu Y, Guo X, Deng R, Liu Y, Feng Y, Mu R, Zhang X, Ma H, Chen Z, Zhang Z, Dong Z, Yang W, Zhang X, Cui Y. Stress dynamically modulates neuronal autophagy to gate depression onset. Nature 2025; 641:427-437. [PMID: 40205038 PMCID: PMC12058529 DOI: 10.1038/s41586-025-08807-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 02/19/2025] [Indexed: 04/11/2025]
Abstract
Chronic stress remodels brain homeostasis, in which persistent change leads to depressive disorders1. As a key modulator of brain homeostasis2, it remains elusive whether and how brain autophagy is engaged in stress dynamics. Here we discover that acute stress activates, whereas chronic stress suppresses, autophagy mainly in the lateral habenula (LHb). Systemic administration of distinct antidepressant drugs similarly restores autophagy function in the LHb, suggesting LHb autophagy as a common antidepressant target. Genetic ablation of LHb neuronal autophagy promotes stress susceptibility, whereas enhancing LHb autophagy exerts rapid antidepressant-like effects. LHb autophagy controls neuronal excitability, synaptic transmission and plasticity by means of on-demand degradation of glutamate receptors. Collectively, this study shows a causal role of LHb autophagy in maintaining emotional homeostasis against stress. Disrupted LHb autophagy is implicated in the maladaptation to chronic stress, and its reversal by autophagy enhancers provides a new antidepressant strategy.
Collapse
Affiliation(s)
- Liang Yang
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Chen Guo
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Zhiwei Zheng
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Yiyan Dong
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qifeng Xie
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Zijian Lv
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Min Li
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yangyang Lu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Xiaonan Guo
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Rongshan Deng
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiqin Liu
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Yirong Feng
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruiqi Mu
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Xuliang Zhang
- Laboratory Animal Center, Zhejiang University, Hangzhou, China
| | - Huan Ma
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Zhong Chen
- Zhejiang Key Laboratory of Neuropsychopharmacology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhijun Zhang
- Department of Neurology, Affiliated Zhongda Hospital, School of Medicine, Institute of Neuropsychiatry, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
- Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhaoqi Dong
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Wei Yang
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiangnan Zhang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
| | - Yihui Cui
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China.
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, Hangzhou, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China.
| |
Collapse
|
3
|
Kochlamazashvili G, Swaminathan A, Stumpf A, Kumar A, Posor Y, Schmitz D, Haucke V, Kuijpers M. Neuronal autophagy controls excitability via ryanodine receptor-mediated regulation of calcium-activated potassium channel function. Proc Natl Acad Sci U S A 2025; 122:e2413651122. [PMID: 40267139 PMCID: PMC12054804 DOI: 10.1073/pnas.2413651122] [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: 07/08/2024] [Accepted: 03/21/2025] [Indexed: 04/25/2025] Open
Abstract
Glutamate-mediated neuronal hyperexcitation plays a causative role in eliciting seizures and promoting epileptogenesis. Recent data suggest that altered autophagy can contribute to the occurrence of epilepsy. We examined the role of autophagy in neuronal physiology by generating knockout mice conditionally lacking the essential autophagy protein ATG5 in glutamatergic neurons. We demonstrate that conditional genetic blockade of neuronal autophagy results in action potential narrowing, axonal hyperexcitability, and an increase in kainate-induced epileptiform bursts ex vivo, indicative of a lower threshold for the induction of epileptic seizures. Neuronal hyperexcitability in hippocampal slices from conditional ATG5 knockout mice is due to elevated activity of the large conductance calcium-activated potassium channel BKCa downstream of calcium influx via the endoplasmic reticulum (ER)-localized calcium channel ryanodine receptor (RYR). Consistently, pharmacological blockade of RYR or BKCa function rescued hyperexcitability and reduced the frequency of kainate-induced epileptiform bursts in ATG5 cKO brain slices. Our findings reveal a physiological role for neuronal autophagy in the regulation of neuronal excitability via the control of RYR-mediated calcium release, and thereby, calcium-activated potassium channel function in the mammalian brain.
Collapse
Affiliation(s)
- Gaga Kochlamazashvili
- Molecular Pharmacology and Cell Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin13125, Germany
| | - Aarti Swaminathan
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin10117, Germany
| | - Alexander Stumpf
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin10117, Germany
| | - Amit Kumar
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin10117, Germany
| | - York Posor
- Molecular Pharmacology and Cell Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin13125, Germany
| | - Dietmar Schmitz
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin10117, Germany
| | - Volker Haucke
- Molecular Pharmacology and Cell Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin13125, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin10117, Germany
- Faculty of Biology, Chemistry, Pharmacy, Freie Universität Berlin, Berlin14195, Germany
| | - Marijn Kuijpers
- Molecular Pharmacology and Cell Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin13125, Germany
- Donders Institute for Brain, Cognition and Behaviour and Faculty of Science, Radboud University, Nijmegen6525AJ, The Netherlands
| |
Collapse
|
4
|
Karpova A, Hiesinger PR, Kuijpers M, Albrecht A, Kirstein J, Andres-Alonso M, Biermeier A, Eickholt BJ, Mikhaylova M, Maglione M, Montenegro-Venegas C, Sigrist SJ, Gundelfinger ED, Haucke V, Kreutz MR. Neuronal autophagy in the control of synapse function. Neuron 2025; 113:974-990. [PMID: 40010347 DOI: 10.1016/j.neuron.2025.01.019] [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: 11/05/2024] [Revised: 12/24/2024] [Accepted: 01/24/2025] [Indexed: 02/28/2025]
Abstract
Neurons are long-lived postmitotic cells that capitalize on autophagy to remove toxic or defective proteins and organelles to maintain neurotransmission and the integrity of their functional proteome. Mutations in autophagy genes cause congenital diseases, sharing prominent brain dysfunctions including epilepsy, intellectual disability, and neurodegeneration. Ablation of core autophagy genes in neurons or glia disrupts normal behavior, leading to motor deficits, memory impairment, altered sociability, and epilepsy, which are associated with defects in synapse maturation, plasticity, and neurotransmitter release. In spite of the importance of autophagy for brain physiology, the substrates of neuronal autophagy and the mechanisms by which defects in autophagy affect synaptic function in health and disease remain controversial. Here, we summarize the current state of knowledge on neuronal autophagy, address the existing controversies and inconsistencies in the field, and provide a roadmap for future research on the role of autophagy in the control of synaptic function.
Collapse
Affiliation(s)
- Anna Karpova
- Leibniz Institute for Neurobiology (LIN), 39118 Magdeburg, Germany; Center for Behavioral Brain Sciences, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | - P Robin Hiesinger
- Faculty of Biology, Chemistry, Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| | - Marijn Kuijpers
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands; Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Anne Albrecht
- Institute of Anatomy, Medical Faculty, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; Center for Behavioral Brain Sciences, Otto-von-Guericke-University, 39120 Magdeburg, Germany; German Center for Mental Health (DZPG), partner site Halle-Jena-Magdeburg, Germany
| | - Janine Kirstein
- Leibniz Institute on Aging-Fritz-Lipmann-Institute, 07754 Jena, Germany; Friedrich-Schiller-Universität, Institute for Biochemistry & Biophysics, 07745 Jena, Germany
| | - Maria Andres-Alonso
- Leibniz Institute for Neurobiology (LIN), 39118 Magdeburg, Germany; Leibniz Group "Dendritic Organelles and Synaptic Function", Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | | | - Britta J Eickholt
- Institute of Molecular Biology and Biochemistry, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Marina Mikhaylova
- Institute of Biology, Humboldt Universität zu Berlin, 10115 Berlin, Germany
| | - Marta Maglione
- Faculty of Biology, Chemistry, Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| | - Carolina Montenegro-Venegas
- Leibniz Institute for Neurobiology (LIN), 39118 Magdeburg, Germany; Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Stephan J Sigrist
- Faculty of Biology, Chemistry, Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany; Institute of Molecular Biology and Biochemistry, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Eckart D Gundelfinger
- Leibniz Institute for Neurobiology (LIN), 39118 Magdeburg, Germany; Center for Behavioral Brain Sciences, Otto-von-Guericke-University, 39120 Magdeburg, Germany; Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Volker Haucke
- Faculty of Biology, Chemistry, Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany; Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Straße 10, 13125 Berlin, Germany; Institute of Molecular Biology and Biochemistry, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany.
| | - Michael R Kreutz
- Leibniz Institute for Neurobiology (LIN), 39118 Magdeburg, Germany; Center for Behavioral Brain Sciences, Otto-von-Guericke-University, 39120 Magdeburg, Germany; Leibniz Group "Dendritic Organelles and Synaptic Function", Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; German Center for Neurodegenerative Diseases (DZNE), Site Magdeburg, 39120 Magdeburg, Germany.
| |
Collapse
|
5
|
Zhu CZ, Li GZ, Lyu HF, Lu YY, Li Y, Zhang XN. Modulation of autophagy by melatonin and its receptors: implications in brain disorders. Acta Pharmacol Sin 2025; 46:525-538. [PMID: 39448859 PMCID: PMC11845611 DOI: 10.1038/s41401-024-01398-2] [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: 06/27/2024] [Accepted: 09/17/2024] [Indexed: 10/26/2024]
Abstract
Autophagy plays a crucial role in maintaining neuronal homeostasis and function, and its disruption is linked to various brain diseases. Melatonin, an endogenous hormone that primarily acts through MT1 and MT2 receptors, regulates autophagy via multiple pathways. Growing evidence indicates that melatonin's ability to modulate autophagy provides therapeutic and preventive benefits in brain disorders, including neurodegenerative and affective diseases. In this review, we summarize the key mechanisms by which melatonin affects autophagy and explore its therapeutic potential in the treatment of brain disorders.
Collapse
Affiliation(s)
- Chen-Ze Zhu
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, 310058, China
| | - Gui-Zhi Li
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 311399, China
| | - Hai-Feng Lyu
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 311399, China
| | - Yang-Yang Lu
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, 310058, China
| | - Yue Li
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, 310058, China
| | - Xiang-Nan Zhang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, 310058, China.
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China.
| |
Collapse
|
6
|
Smith EM, Coughlan ML, Maday S. Turning garbage into gold: Autophagy in synaptic function. Curr Opin Neurobiol 2025; 90:102937. [PMID: 39667255 PMCID: PMC11903044 DOI: 10.1016/j.conb.2024.102937] [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: 06/02/2024] [Revised: 10/26/2024] [Accepted: 11/13/2024] [Indexed: 12/14/2024]
Abstract
Trillions of synapses in the human brain enable thought and behavior. Synaptic connections must be established and maintained, while retaining dynamic flexibility to respond to experiences. These processes require active remodeling of the synapse to control the composition and integrity of proteins and organelles. Macroautophagy (hereafter, autophagy) provides a mechanism to edit and prune the synaptic proteome. Canonically, autophagy has been viewed as a homeostatic process, which eliminates aged and damaged proteins to maintain neuronal survival. However, accumulating evidence suggests that autophagy also degrades specific cargoes in response to neuronal activity to impact neuronal transmission, excitability, and synaptic plasticity. Here, we will discuss the diverse roles, regulation, and mechanisms of neuronal autophagy in synaptic function and contributions from glial autophagy in these processes.
Collapse
Affiliation(s)
- Erin Marie Smith
- Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maeve Louise Coughlan
- Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sandra Maday
- Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
7
|
Tutas J, Tolve M, Özer-Yildiz E, Ickert L, Klein I, Silverman Q, Liebsch F, Dethloff F, Giavalisco P, Endepols H, Georgomanolis T, Neumaier B, Drzezga A, Schwarz G, Thorens B, Gatto G, Frezza C, Kononenko NL. Autophagy regulator ATG5 preserves cerebellar function by safeguarding its glycolytic activity. Nat Metab 2025; 7:297-320. [PMID: 39815080 PMCID: PMC11860254 DOI: 10.1038/s42255-024-01196-4] [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: 02/02/2024] [Accepted: 11/29/2024] [Indexed: 01/18/2025]
Abstract
Dysfunctions in autophagy, a cellular mechanism for breaking down components within lysosomes, often lead to neurodegeneration. The specific mechanisms underlying neuronal vulnerability due to autophagy dysfunction remain elusive. Here we show that autophagy contributes to cerebellar Purkinje cell (PC) survival by safeguarding their glycolytic activity. Outside the conventional housekeeping role, autophagy is also involved in the ATG5-mediated regulation of glucose transporter 2 (GLUT2) levels during cerebellar maturation. Autophagy-deficient PCs exhibit GLUT2 accumulation on the plasma membrane, along with increased glucose uptake and alterations in glycolysis. We identify lysophosphatidic acid and serine as glycolytic intermediates that trigger PC death and demonstrate that the deletion of GLUT2 in ATG5-deficient mice mitigates PC neurodegeneration and rescues their ataxic gait. Taken together, this work reveals a mechanism for regulating GLUT2 levels in neurons and provides insights into the neuroprotective role of autophagy by controlling glucose homeostasis in the brain.
Collapse
Affiliation(s)
- Janine Tutas
- CECAD Excellence Center, University of Cologne, Cologne, Germany
- Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Marianna Tolve
- CECAD Excellence Center, University of Cologne, Cologne, Germany
- Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Ebru Özer-Yildiz
- CECAD Excellence Center, University of Cologne, Cologne, Germany
- Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Lotte Ickert
- CECAD Excellence Center, University of Cologne, Cologne, Germany
- Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Ines Klein
- Department of Neurology, University Hospital of Cologne, Cologne, Germany
| | - Quinn Silverman
- Department of Neurology, University Hospital of Cologne, Cologne, Germany
| | - Filip Liebsch
- Institute of Biochemistry, Department of Chemistry, University of Cologne, Cologne, Germany
| | | | | | - Heike Endepols
- Department of Nuclear Medicine, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Jülich, Germany
| | | | - Bernd Neumaier
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Jülich, Germany
| | - Alexander Drzezga
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Molecular Organization of the Brain (INM-2), Jülich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn-Cologne, Germany
| | - Guenter Schwarz
- Institute of Biochemistry, Department of Chemistry, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Bernard Thorens
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Graziana Gatto
- Department of Neurology, University Hospital of Cologne, Cologne, Germany
| | - Christian Frezza
- CECAD Excellence Center, University of Cologne, Cologne, Germany
- Institute for Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Natalia L Kononenko
- CECAD Excellence Center, University of Cologne, Cologne, Germany.
- Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
- Institute for Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany.
| |
Collapse
|
8
|
Ham A, Chang AY, Li H, Bain JM, Goldman JE, Sulzer D, Veenstra-VanderWeele J, Tang G. Impaired macroautophagy confers substantial risk for intellectual disability in children with autism spectrum disorders. Mol Psychiatry 2025; 30:810-824. [PMID: 39237724 DOI: 10.1038/s41380-024-02741-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Autism spectrum disorder (ASD) represents a complex of neurological and developmental disabilities characterized by clinical and genetic heterogeneity. While the causes of ASD are still unknown, many ASD risk factors are found to converge on intracellular quality control mechanisms that are essential for cellular homeostasis, including the autophagy-lysosomal degradation pathway. Studies have reported impaired autophagy in ASD human brain and ASD-like synapse pathology and behaviors in mouse models of brain autophagy deficiency, highlighting an essential role for defective autophagy in ASD pathogenesis. To determine whether altered autophagy in the brain may also occur in peripheral cells that might provide useful biomarkers, we assessed activities of autophagy in lympoblasts from ASD and control subjects. We find that lymphoblast autophagy is compromised in a subset of ASD participants due to impaired autophagy induction. Similar changes in autophagy are detected in postmortem human brains from ASD individuals and in brain and peripheral blood mononuclear cells from syndromic ASD mouse models. Remarkably, we find a strong correlation between impaired autophagy and intellectual disability in ASD participants. By depleting the key autophagy gene Atg7 from different brain cells, we provide further evidence that autophagy deficiency causes cognitive impairment in mice. Together, our findings suggest autophagy dysfunction as a convergent mechanism that can be detected in peripheral blood cells from a subset of autistic individuals, and that lymphoblast autophagy may serve as a biomarker to stratify ASD patients for the development of targeted interventions.
Collapse
Affiliation(s)
- Ahrom Ham
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Audrey Yuen Chang
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Hongyu Li
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Jennifer M Bain
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - James E Goldman
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - David Sulzer
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Pharmacology, Columbia University Irving Medical Center, New York, NY, 10032, USA
- New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Jeremy Veenstra-VanderWeele
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, 10032, USA
- New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Guomei Tang
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
9
|
Jahanian S, Gulbronson CI, Gransee HM, Millesi E, Sieck GC, Mantilla CB. Chloroquine Affects Presynaptic Membrane Retrieval in Diaphragm Neuromuscular Junctions of Old Mice. Int J Mol Sci 2024; 26:43. [PMID: 39795904 PMCID: PMC11719459 DOI: 10.3390/ijms26010043] [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: 12/11/2024] [Accepted: 12/18/2024] [Indexed: 01/13/2025] Open
Abstract
Aging disrupts multiple homeostatic processes, including autophagy, a cellular process for the recycling and degradation of defective cytoplasmic structures. Acute treatment with the autophagy inhibitor chloroquine blunts the maximal forces generated by the diaphragm muscle, but the mechanisms underlying neuromuscular dysfunction in old age remain poorly understood. We hypothesized that chloroquine treatment increases the presynaptic retention of the styryl dye FM 4-64 following high-frequency nerve stimulation, consistent with the accumulation of unprocessed bulk endosomes. Diaphragm-phrenic nerve preparations from 24-month-old male and female C57BL/6 × 129 J mice were incubated with FM 4-64 (5 µM) and either chloroquine (50 µM) or vehicle during 80 Hz phrenic nerve stimulation. Acute chloroquine treatment significantly decreased FM 4-64 intensity at diaphragm neuromuscular junctions following 80 Hz phrenic nerve stimulation, consistent with disrupted synaptic vesicle recycling. A similar reduction was evident in regions with the greatest FM 4-64 fluorescence intensity, which most likely surround synaptic vesicle release sites. In the absence of nerve stimulation, chloroquine treatment significantly increased FM 4-64 intensity at diaphragm neuromuscular junctions. These findings highlight the importance of autophagy in regulating presynaptic vesicle retrieval (including vesicle recycling and endosomal processing) and support the role of autophagy impairments in age-related neuromuscular dysfunction.
Collapse
Affiliation(s)
- Sepideh Jahanian
- Department of Anesthesiology & Perioperative Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Chloe I. Gulbronson
- Department of Anesthesiology & Perioperative Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Heather M. Gransee
- Department of Anesthesiology & Perioperative Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Elena Millesi
- Department of Surgery Research Services, Mayo Clinic, Rochester, MN 55905, USA
| | - Gary C. Sieck
- Department of Anesthesiology & Perioperative Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Carlos B. Mantilla
- Department of Anesthesiology & Perioperative Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
10
|
Salvatore SV, Ilagan MXG, Shu H, Lambert PM, Benz A, Qian M, Covey DF, Zorumski CF, Mennerick S. Neuroactive steroid effects on autophagy in a human embryonic kidney 293 (HEK) cell model. Sci Rep 2024; 14:1042. [PMID: 38200205 PMCID: PMC10781668 DOI: 10.1038/s41598-024-51582-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: 07/18/2023] [Accepted: 01/07/2024] [Indexed: 01/12/2024] Open
Abstract
Neuropsychiatric and neurodegenerative disorders are correlated with cellular stress. Macroautophagy (autophagy) may represent an important protective pathway to maintain cellular homeostasis and functionality, as it targets cytoplasmic components to lysosomes for degradation and recycling. Given recent evidence that some novel psychiatric treatments, such as the neuroactive steroid (NAS) allopregnanolone (AlloP, brexanolone), may induce autophagy, we stably transfected human embryonic kidney 293 (HEK) cells with a ratiometric fluorescent probe to assay NAS effects on autophagy. We hypothesized that NAS may modulate autophagy in part by the ability of uncharged NAS to readily permeate membranes. Microscopy revealed a weak effect of AlloP on autophagic flux compared with the positive control treatment of Torin1. In high-throughput microplate experiments, we found that autophagy induction was more robust in early passages of HEK cells. Despite limiting studies to early passages for maximum sensitivity, a range of NAS structures failed to reliably induce autophagy or interact with Torin1 or starvation effects. To probe NAS in a system where AlloP effects have been shown previously, we surveyed astrocytes and again saw minimal autophagy induction by AlloP. Combined with other published results, our results suggest that NAS may modulate autophagy in a cell-specific or context-specific manner. Although there is merit to cell lines as a screening tool, future studies may require assaying NAS in cells from brain regions involved in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sofia V Salvatore
- Departments of Psychiatry, Washington University in St. Louis School of Medicine, 660 S. Euclid Ave., MSC 8134-0181-0G, St. Louis, MO, 63110, USA
| | - Ma Xenia G Ilagan
- High-Throughput Screening Core, Center for Drug Discovery, Washington University in St. Louis School of Medicine, 660 S. Euclid Ave., MSC 8134-0181-0G, St. Louis, MO, 63110, USA
| | - Hongjin Shu
- Departments of Psychiatry, Washington University in St. Louis School of Medicine, 660 S. Euclid Ave., MSC 8134-0181-0G, St. Louis, MO, 63110, USA
| | - Peter M Lambert
- Departments of Psychiatry, Washington University in St. Louis School of Medicine, 660 S. Euclid Ave., MSC 8134-0181-0G, St. Louis, MO, 63110, USA
- Medical Scientist Training Program, Washington University in St. Louis School of Medicine, 660 S. Euclid Ave., MSC 8134-0181-0G, St. Louis, MO, 63110, USA
| | - Ann Benz
- Departments of Psychiatry, Washington University in St. Louis School of Medicine, 660 S. Euclid Ave., MSC 8134-0181-0G, St. Louis, MO, 63110, USA
| | - Mingxing Qian
- Developmental Biology, Washington University in St. Louis School of Medicine, 660 S. Euclid Ave., MSC 8134-0181-0G, St. Louis, MO, 63110, USA
| | - Douglas F Covey
- Developmental Biology, Washington University in St. Louis School of Medicine, 660 S. Euclid Ave., MSC 8134-0181-0G, St. Louis, MO, 63110, USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis School of Medicine, 660 S. Euclid Ave., MSC 8134-0181-0G, St. Louis, MO, 63110, USA
| | - Charles F Zorumski
- Departments of Psychiatry, Washington University in St. Louis School of Medicine, 660 S. Euclid Ave., MSC 8134-0181-0G, St. Louis, MO, 63110, USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis School of Medicine, 660 S. Euclid Ave., MSC 8134-0181-0G, St. Louis, MO, 63110, USA
| | - Steven Mennerick
- Departments of Psychiatry, Washington University in St. Louis School of Medicine, 660 S. Euclid Ave., MSC 8134-0181-0G, St. Louis, MO, 63110, USA.
- Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis School of Medicine, 660 S. Euclid Ave., MSC 8134-0181-0G, St. Louis, MO, 63110, USA.
| |
Collapse
|
11
|
Mabuchi Y, Cui X, Xie L, Kim H, Jiang T, Yapici N. GABA-mediated inhibition in visual feedback neurons fine-tunes Drosophila male courtship. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.25.525544. [PMID: 36747836 PMCID: PMC9900824 DOI: 10.1101/2023.01.25.525544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Vision is critical for the regulation of mating behaviors in many species. Here, we discovered that the Drosophila ortholog of human GABA A -receptor-associated protein (GABARAP) is required to fine-tune male courtship by modulating the activity of visual feedback neurons, lamina tangential cells (Lat). GABARAP is a ubiquitin-like protein that regulates cell-surface levels of GABA A receptors. Knocking down GABARAP or GABA A receptors in Lat neurons or hyperactivating them induces male courtship toward other males. Inhibiting Lat neurons, on the other hand, delays copulation by impairing the ability of males to follow females. Remarkably, the human ortholog of Drosophila GABARAP restores function in Lat neurons. Using in vivo two-photon imaging and optogenetics, we show that Lat neurons are functionally connected to neural circuits that mediate visually-guided courtship pursuits in males. Our work reveals a novel physiological role for GABARAP in fine-tuning the activity of a visual circuit that tracks a mating partner during courtship.
Collapse
Affiliation(s)
- Yuta Mabuchi
- Department of Neurobiology and Behavior, Cornell University, 14853, Ithaca, NY, USA
| | - Xinyue Cui
- Department of Neurobiology and Behavior, Cornell University, 14853, Ithaca, NY, USA
| | - Lily Xie
- Department of Neurobiology and Behavior, Cornell University, 14853, Ithaca, NY, USA
| | - Haein Kim
- Department of Neurobiology and Behavior, Cornell University, 14853, Ithaca, NY, USA
| | - Tianxing Jiang
- Department of Neurobiology and Behavior, Cornell University, 14853, Ithaca, NY, USA
| | - Nilay Yapici
- Department of Neurobiology and Behavior, Cornell University, 14853, Ithaca, NY, USA
| |
Collapse
|
12
|
Baratta AM, Mangieri RA, Aziz HC, Lopez MF, Farris SP, Homanics GE. Effect of chronic intermittent ethanol vapor exposure on RNA content of brain-derived extracellular vesicles. Alcohol 2022; 105:9-24. [PMID: 36055466 PMCID: PMC10173183 DOI: 10.1016/j.alcohol.2022.08.006] [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: 03/17/2022] [Revised: 08/18/2022] [Accepted: 08/18/2022] [Indexed: 01/26/2023]
Abstract
Extracellular vesicles (EVs) are important players in normal biological function and disease pathogenesis. Of the many biomolecules packaged into EVs, coding and noncoding RNA transcripts are of particular interest for their ability to significantly alter cellular and molecular processes. Here we investigate how chronic ethanol exposure impacts EV RNA cargo and the functional outcomes of these changes. Following chronic intermittent ethanol (CIE) vapor exposure, EVs were isolated from male and female C57BL/6J mouse brain. Total RNA from EVs was analyzed by lncRNA/mRNA microarray to survey changes in RNA cargo following vapor exposure. Differential expression analysis of microarray data revealed a number of lncRNA and mRNA types differentially expressed in CIE compared to control EVs. Weighted gene co-expression network analysis identified multiple male and female specific modules related to neuroinflammation, cell death, demyelination, and synapse organization. To functionally test these changes, whole-cell voltage-clamp recordings were used to assess synaptic transmission. Incubation of nucleus accumbens brain slices with EVs led to a reduction in spontaneous excitatory postsynaptic current amplitude, although no changes in synaptic transmission were observed between control and CIE EV administration. These results indicate that CIE vapor exposure significantly changes the RNA cargo of brain-derived EVs, which have the ability to impact neuronal function.
Collapse
Affiliation(s)
- Annalisa M Baratta
- Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Regina A Mangieri
- College of Pharmacy, University of Texas at Austin, Texas, United States
| | - Heather C Aziz
- College of Pharmacy, University of Texas at Austin, Texas, United States
| | - Marcelo F Lopez
- Department of Psychiatry and Behavioral Science, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Sean P Farris
- Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States; Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States; Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Gregg E Homanics
- Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States; Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States.
| |
Collapse
|
13
|
McGuirt A, Pigulevskiy I, Sulzer D. Developmental regulation of thalamus-driven pauses in striatal cholinergic interneurons. iScience 2022; 25:105332. [PMID: 36325074 PMCID: PMC9619292 DOI: 10.1016/j.isci.2022.105332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 08/18/2022] [Accepted: 10/10/2022] [Indexed: 01/24/2023] Open
Abstract
In response to salient sensory cues, the tonically active striatal cholinergic interneuron (ChI) exhibits a characteristic synchronized "pause" thought to facilitate learning and the execution of motivated behavior. We report that thalamostriatal-driven ChI pauses are enhanced in ex vivo brain slices from infantile (P10) mice, with decreasing expression in preadolescent (P28) and adult (P100) mice concurrent with waning excitatory input to ChIs. Our data are consistent with previous reports that the adult ChI pause is dependent on dopamine signaling, but we find that the robust pausing at P10 is dopamine independent. Instead, elevated expression of the noninactivating delayed rectifier Kv7.2/3 current promotes pausing in infantile ChIs. Because this current decreases over development, a parallel increase in Ih further attenuates pause expression. These findings demonstrate that cell intrinsic and circuit mechanisms of ChI pause expression are developmentally determined and may underlie changes in learning properties as the nervous system matures.
Collapse
Affiliation(s)
- Avery McGuirt
- Departments of Psychiatry, Neurology, Pharmacology, Columbia University Irving Medical Center, Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Irena Pigulevskiy
- Departments of Psychiatry, Neurology, Pharmacology, Columbia University Irving Medical Center, Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - David Sulzer
- Departments of Psychiatry, Neurology, Pharmacology, Columbia University Irving Medical Center, Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| |
Collapse
|
14
|
Choi I, Heaton GR, Lee YK, Yue Z. Regulation of α-synuclein homeostasis and inflammasome activation by microglial autophagy. SCIENCE ADVANCES 2022; 8:eabn1298. [PMID: 36288297 PMCID: PMC9604594 DOI: 10.1126/sciadv.abn1298] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 07/11/2022] [Indexed: 05/07/2023]
Abstract
Autophagy clears protein aggregates, damaged cellular organelles, and pathogens through the lysosome. Although autophagy is highly conserved across all cell types, its activity in each cell is specifically adapted to carry out distinct physiological functions. The role of autophagy in neurons has been well characterized; however, in glial cells, its function remains largely unknown. Microglia are brain-resident macrophages that survey the brain to remove injured neurons, excessive synapses, protein aggregates, and infectious agents. Current studies have demonstrated that dysfunctional microglia contribute to neurodegenerative diseases. In Alzheimer's disease animal models, microglia play a critical role in regulating amyloid plaque formation and neurotoxicity. However, how microglia are involved in Parkinson's disease (PD) remains poorly understood. Propagation of aggregated α-synuclein via cell-to-cell transmission and neuroinflammation have emerged as important mechanisms underlying neuropathologies in PD. Here, we review converging evidence that microglial autophagy maintains α-synuclein homeostasis, regulates neuroinflammation, and confers neuroprotection in PD experimental models.
Collapse
Affiliation(s)
| | - George R. Heaton
- Department of Neurology and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - You-Kyung Lee
- Department of Neurology and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | |
Collapse
|
15
|
Abstract
Macroautophagy is an evolutionarily conserved process that delivers diverse cellular contents to lysosomes for degradation. As our understanding of this pathway grows, so does our appreciation for its importance in disorders of the CNS. Once implicated primarily in neurodegenerative events owing to acute injury and ageing, macroautophagy is now also linked to disorders of neurodevelopment, indicating that it is essential for both the formation and maintenance of a healthy CNS. In parallel to understanding the significance of macroautophagy across contexts, we have gained a greater mechanistic insight into its physiological regulation and the breadth of cargoes it can degrade. Macroautophagy is a broadly used homeostatic process, giving rise to questions surrounding how defects in this single pathway could cause diseases with distinct clinical and pathological signatures. To address this complexity, we herein review macroautophagy in the mammalian CNS by examining three key features of the process and its relationship to disease: how it functions at a basal level in the discrete cell types of the brain and spinal cord; which cargoes are being degraded in physiological and pathological settings; and how the different stages of the macroautophagy pathway intersect with diseases of neurodevelopment and adult-onset neurodegeneration.
Collapse
Affiliation(s)
- Christopher J Griffey
- Doctoral Program in Neurobiology and Behaviour, Medical Scientist Training Program, Columbia University, New York, NY, USA
| | - Ai Yamamoto
- Departments of Neurology, and Pathology and Cell Biology, Columbia University, New York, NY, USA.
| |
Collapse
|
16
|
Kallergi E, Daskalaki AD, Kolaxi A, Camus C, Ioannou E, Mercaldo V, Haberkant P, Stein F, Sidiropoulou K, Dalezios Y, Savitski MM, Bagni C, Choquet D, Hosy E, Nikoletopoulou V. Dendritic autophagy degrades postsynaptic proteins and is required for long-term synaptic depression in mice. Nat Commun 2022; 13:680. [PMID: 35115539 PMCID: PMC8814153 DOI: 10.1038/s41467-022-28301-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/14/2022] [Indexed: 01/18/2023] Open
Abstract
The pruning of dendritic spines during development requires autophagy. This process is facilitated by long-term depression (LTD)-like mechanisms, which has led to speculation that LTD, a fundamental form of synaptic plasticity, also requires autophagy. Here, we show that the induction of LTD via activation of NMDA receptors or metabotropic glutamate receptors initiates autophagy in the postsynaptic dendrites in mice. Dendritic autophagic vesicles (AVs) act in parallel with the endocytic machinery to remove AMPA receptor subunits from the membrane for degradation. During NMDAR-LTD, key postsynaptic proteins are sequestered for autophagic degradation, as revealed by quantitative proteomic profiling of purified AVs. Pharmacological inhibition of AV biogenesis, or conditional ablation of atg5 in pyramidal neurons abolishes LTD and triggers sustained potentiation in the hippocampus. These deficits in synaptic plasticity are recapitulated by knockdown of atg5 specifically in postsynaptic pyramidal neurons in the CA1 area. Conducive to the role of synaptic plasticity in behavioral flexibility, mice with autophagy deficiency in excitatory neurons exhibit altered response in reversal learning. Therefore, local assembly of the autophagic machinery in dendrites ensures the degradation of postsynaptic components and facilitates LTD expression.
Collapse
Affiliation(s)
- Emmanouela Kallergi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, 1005, Switzerland
| | | | - Angeliki Kolaxi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, 1005, Switzerland
| | - Come Camus
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Evangelia Ioannou
- School of Biological Sciences, University of Crete, Heraklion, 70013, Greece
| | - Valentina Mercaldo
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, 1005, Switzerland
| | - Per Haberkant
- Proteomic Core Facility (PCF), European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Frank Stein
- Proteomic Core Facility (PCF), European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | | | - Yannis Dalezios
- School of Medicine, University of Crete, Heraklion, 71003, Greece
- Institute of Applied and Computational Mathematics (IACM), Foundation for Research and Technology-Hellas (FORTH), Heraklion, Greece
| | - Mikhail M Savitski
- Proteomic Core Facility (PCF), European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), University of Rome Tor Vergata, Rome, 00133, Italy
| | - Claudia Bagni
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, 1005, Switzerland
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, 00133, Italy
| | - Daniel Choquet
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France
- University of Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4, F-33000, Bordeaux, France
| | - Eric Hosy
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France
| | | |
Collapse
|
17
|
Systemic LPS-induced microglial activation results in increased GABAergic tone: A mechanism of protection against neuroinflammation in the medial prefrontal cortex in mice. Brain Behav Immun 2022; 99:53-69. [PMID: 34582995 DOI: 10.1016/j.bbi.2021.09.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/19/2021] [Accepted: 09/21/2021] [Indexed: 02/01/2023] Open
Abstract
Neuroinflammation with excess microglial activation and synaptic dysfunction are early symptoms of most neurological diseases. However, how microglia-associated neuroinflammation regulates synaptic activity remains obscure. We report here that acute neuroinflammation induced by intraperitoneal injection of lipopolysaccharide (LPS) results in cell-type-specific increases in inhibitory postsynaptic currents in the glutamatergic, but not the GABAergic, neurons of medial prefrontal cortex (mPFC), coinciding with excessive microglial activation. LPS causes upregulation in levels of GABAAR subunits, glutamine synthetase and vesicular GABA transporter, and downregulation in brain-derived neurotrophic factor (BDNF) and its receptor, pTrkB. Blockage of microglial activation by minocycline ameliorates LPS-induced abnormal expression of GABA signaling-related proteins and activity of synaptic and network. Moreover, minocycline prevents the mice from LPS-induced aberrant behavior, such as a reduction in total distance and time spent in the centre in the open field test; decreases in entries into the open arm of elevated-plus maze and in consumption of sucrose; increased immobility in the tail suspension test. Furthermore, upregulation of GABA signaling by tiagabine also prevents LPS-induced microglial activation and aberrant behavior. This study illustrates a mode of bidirectional constitutive signaling between the neural and immune compartments of the brain, and suggests that the mPFC is an important area for brain-immune system communication. Moreover, the present study highlights GABAergic signaling as a key therapeutic target for mitigating neuroinflammation-induced abnormal synaptic activity in the mPFC, together with the associated behavioral abnormalities.
Collapse
|
18
|
Pagani M, Barsotti N, Bertero A, Trakoshis S, Ulysse L, Locarno A, Miseviciute I, De Felice A, Canella C, Supekar K, Galbusera A, Menon V, Tonini R, Deco G, Lombardo MV, Pasqualetti M, Gozzi A. mTOR-related synaptic pathology causes autism spectrum disorder-associated functional hyperconnectivity. Nat Commun 2021; 12:6084. [PMID: 34667149 PMCID: PMC8526836 DOI: 10.1038/s41467-021-26131-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 09/17/2021] [Indexed: 11/24/2022] Open
Abstract
Postmortem studies have revealed increased density of excitatory synapses in the brains of individuals with autism spectrum disorder (ASD), with a putative link to aberrant mTOR-dependent synaptic pruning. ASD is also characterized by atypical macroscale functional connectivity as measured with resting-state fMRI (rsfMRI). These observations raise the question of whether excess of synapses causes aberrant functional connectivity in ASD. Using rsfMRI, electrophysiology and in silico modelling in Tsc2 haploinsufficient mice, we show that mTOR-dependent increased spine density is associated with ASD -like stereotypies and cortico-striatal hyperconnectivity. These deficits are completely rescued by pharmacological inhibition of mTOR. Notably, we further demonstrate that children with idiopathic ASD exhibit analogous cortical-striatal hyperconnectivity, and document that this connectivity fingerprint is enriched for ASD-dysregulated genes interacting with mTOR or Tsc2. Finally, we show that the identified transcriptomic signature is predominantly expressed in a subset of children with autism, thereby defining a segregable autism subtype. Our findings causally link mTOR-related synaptic pathology to large-scale network aberrations, revealing a unifying multi-scale framework that mechanistically reconciles developmental synaptopathy and functional hyperconnectivity in autism.
Collapse
Affiliation(s)
- Marco Pagani
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
- Autism Center, Child Mind Institute, New York, NY, USA
| | - Noemi Barsotti
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa, Italy
| | - Alice Bertero
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa, Italy
| | - Stavros Trakoshis
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
- Laboratory for Autism and Neurodevelopmental Disorders, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
| | - Laura Ulysse
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Barcelona, Spain
| | - Andrea Locarno
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| | - Ieva Miseviciute
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| | - Alessia De Felice
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
| | - Carola Canella
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
| | | | - Alberto Galbusera
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
| | | | - Raffaella Tonini
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| | - Gustavo Deco
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Barcelona, Spain
- Institució Catalana de la Recerca i Estudis Avançats (ICREA), Universitat Pompeu Fabra, Barcelona, Spain
| | - Michael V Lombardo
- Laboratory for Autism and Neurodevelopmental Disorders, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
- Autism Research Centre, University of Cambridge, Cambridge, UK
| | - Massimo Pasqualetti
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa, Italy
| | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy.
| |
Collapse
|
19
|
Fernandes HJR, Patikas N, Foskolou S, Field SF, Park JE, Byrne ML, Bassett AR, Metzakopian E. Single-Cell Transcriptomics of Parkinson's Disease Human In Vitro Models Reveals Dopamine Neuron-Specific Stress Responses. Cell Rep 2021; 33:108263. [PMID: 33053338 DOI: 10.1016/j.celrep.2020.108263] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/29/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022] Open
Abstract
The advent of induced pluripotent stem cell (iPSC)-derived neurons has revolutionized Parkinson's disease (PD) research, but single-cell transcriptomic analysis suggests unresolved cellular heterogeneity within these models. Here, we perform the largest single-cell transcriptomic study of human iPSC-derived dopaminergic neurons to elucidate gene expression dynamics in response to cytotoxic and genetic stressors. We identify multiple neuronal subtypes with transcriptionally distinct profiles and differential sensitivity to stress, highlighting cellular heterogeneity in dopamine in vitro models. We validate this disease model by showing robust expression of PD GWAS genes and overlap with postmortem adult substantia nigra neurons. Importantly, stress signatures are ameliorated using felodipine, an FDA-approved drug. Using isogenic SNCA-A53T mutants, we find perturbations in glycolysis, cholesterol metabolism, synaptic signaling, and ubiquitin-proteasomal degradation. Overall, our study reveals cell type-specific perturbations in human dopamine neurons, which will further our understanding of PD and have implications for cell replacement therapies.
Collapse
Affiliation(s)
- Hugo J R Fernandes
- UK Dementia Research Institute, Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AH, UK; Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Nikolaos Patikas
- UK Dementia Research Institute, Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AH, UK
| | - Stefanie Foskolou
- UK Dementia Research Institute, Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AH, UK; Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Sarah F Field
- UK Dementia Research Institute, Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AH, UK
| | - Jong-Eun Park
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK; Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Meg L Byrne
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Andrew R Bassett
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Emmanouil Metzakopian
- UK Dementia Research Institute, Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AH, UK.
| |
Collapse
|
20
|
Sumitomo A, Tomoda T. Autophagy in neuronal physiology and disease. Curr Opin Pharmacol 2021; 60:133-140. [PMID: 34416525 DOI: 10.1016/j.coph.2021.07.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/04/2021] [Accepted: 07/13/2021] [Indexed: 12/21/2022]
Abstract
Neural circuit functions critically depend on homeostatic regulation and quality control of neuronal proteins and organelles. Emerging evidence shows that autophagy, cellular clearance machinery, selectively degrades or controls homeostasis of both pre- and post-synaptic components (e.g. synaptic proteins, organelles, neurotransmitters, and their receptors), thereby regulating synaptic remodeling, neurotransmission, and neuroplasticity. Along with its well-known role in supporting neuronal cell viability and neurodevelopment, autophagy is now implicated in a wide range of neuronal physiology throughout neuronal lifetime, including higher-order brain functions such as information processing, memory encoding, or cognitive functions. Here, we review recent literature on the roles of neuronal autophagy in homeostatic maintenance of synaptic functions and discuss how disruptions in these processes may contribute to the pathophysiology of neurodevelopmental and psychiatric disorders.
Collapse
Affiliation(s)
- Akiko Sumitomo
- Centre for Addiction and Mental Health, Toronto, Ontario M5T 1R8, Canada
| | - Toshifumi Tomoda
- Centre for Addiction and Mental Health, Toronto, Ontario M5T 1R8, Canada.
| |
Collapse
|
21
|
Pan PY, Zhu J, Rizvi A, Zhu X, Tanaka H, Dreyfus CF. Synaptojanin1 deficiency upregulates basal autophagosome formation in astrocytes. J Biol Chem 2021; 297:100873. [PMID: 34126070 PMCID: PMC8258991 DOI: 10.1016/j.jbc.2021.100873] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/02/2021] [Accepted: 06/10/2021] [Indexed: 01/07/2023] Open
Abstract
Macroautophagy dysregulation is implicated in multiple neurological disorders, such as Parkinson's disease. While autophagy pathways are heavily researched in heterologous cells and neurons, regulation of autophagy in the astrocyte, the most abundant cell type in the mammalian brain, is less well understood. Missense mutations in the Synj1 gene encoding Synaptojanin1 (Synj1), a neuron-enriched lipid phosphatase, have been linked to Parkinsonism with seizures. Our previous study showed that the Synj1 haploinsufficient (Synj1+/−) mouse exhibits age-dependent autophagy impairment in multiple brain regions. Here, we used cultured astrocytes from Synj1-deficient mice to investigate its role in astrocyte autophagy. We report that Synj1 is expressed in low levels in astrocytes and represses basal autophagosome formation. We demonstrate using cellular imaging that Synj1-deficient astrocytes exhibit hyperactive autophagosome formation, represented by an increase in the size and number of GFP-microtubule-associated protein 1A/1B-light chain 3 structures. Interestingly, Synj1 deficiency is also associated with an impairment in stress-induced autophagy clearance. We show, for the first time, that the Parkinsonism-associated R839C mutation impacts autophagy in astrocytes. The impact of this mutation on the phosphatase function of Synj1 resulted in elevated basal autophagosome formation that mimics Synj1 deletion. We found that the membrane expression of the astrocyte-specific glucose transporter GluT-1 was reduced in Synj1-deficient astrocytes. Consistently, AMP-activated protein kinase activity was elevated, suggesting altered glucose sensing in Synj1-deficient astrocytes. Expressing exogenous GluT-1 in Synj1-deficient astrocytes reversed the autophagy impairment, supporting a role for Synj1 in regulating astrocyte autophagy via disrupting glucose-sensing pathways. Thus, our work suggests a novel mechanism for Synj1-related Parkinsonism involving astrocyte dysfunction.
Collapse
Affiliation(s)
- Ping-Yue Pan
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA.
| | - Justin Zhu
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Asma Rizvi
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Xinyu Zhu
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Hikari Tanaka
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Cheryl F Dreyfus
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
22
|
Bedont JL, Toda H, Shi M, Park CH, Quake C, Stein C, Kolesnik A, Sehgal A. Short and long sleeping mutants reveal links between sleep and macroautophagy. eLife 2021; 10:64140. [PMID: 34085929 PMCID: PMC8177895 DOI: 10.7554/elife.64140] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 05/20/2021] [Indexed: 02/03/2023] Open
Abstract
Sleep is a conserved and essential behavior, but its mechanistic and functional underpinnings remain poorly defined. Through unbiased genetic screening in Drosophila, we discovered a novel short-sleep mutant we named argus. Positional cloning and subsequent complementation, CRISPR/Cas9 knock-out, and RNAi studies identified Argus as a transmembrane protein that acts in adult peptidergic neurons to regulate sleep. argus mutants accumulate undigested Atg8a(+) autophagosomes, and genetic manipulations impeding autophagosome formation suppress argus sleep phenotypes, indicating that autophagosome accumulation drives argus short-sleep. Conversely, a blue cheese neurodegenerative mutant that impairs autophagosome formation was identified independently as a gain-of-sleep mutant, and targeted RNAi screens identified additional genes involved in autophagosome formation whose knockdown increases sleep. Finally, autophagosomes normally accumulate during the daytime and nighttime sleep deprivation extends this accumulation into the following morning, while daytime gaboxadol feeding promotes sleep and reduces autophagosome accumulation at nightfall. In sum, our results paradoxically demonstrate that wakefulness increases and sleep decreases autophagosome levels under unperturbed conditions, yet strong and sustained upregulation of autophagosomes decreases sleep, whereas strong and sustained downregulation of autophagosomes increases sleep. The complex relationship between sleep and autophagy suggested by our findings may have implications for pathological states including chronic sleep disorders and neurodegeneration, as well as for integration of sleep need with other homeostats, such as under conditions of starvation.
Collapse
Affiliation(s)
- Joseph L Bedont
- Chronobiology and Sleep Institute, Perelman Medical School of University of Pennsylvania, Philadelphia, United States
| | - Hirofumi Toda
- Chronobiology and Sleep Institute, Perelman Medical School of University of Pennsylvania, Philadelphia, United States
| | - Mi Shi
- Chronobiology and Sleep Institute, Perelman Medical School of University of Pennsylvania, Philadelphia, United States
| | - Christine H Park
- Chronobiology and Sleep Institute, Perelman Medical School of University of Pennsylvania, Philadelphia, United States
| | - Christine Quake
- Chronobiology and Sleep Institute, Perelman Medical School of University of Pennsylvania, Philadelphia, United States
| | - Carly Stein
- Chronobiology and Sleep Institute, Perelman Medical School of University of Pennsylvania, Philadelphia, United States
| | - Anna Kolesnik
- Chronobiology and Sleep Institute, Perelman Medical School of University of Pennsylvania, Philadelphia, United States
| | - Amita Sehgal
- Chronobiology and Sleep Institute, Perelman Medical School of University of Pennsylvania, Philadelphia, United States.,Howard Hughes Medical Institute, Philadelphia, United States
| |
Collapse
|
23
|
La Barbera L, Vedele F, Nobili A, Krashia P, Spoleti E, Latagliata EC, Cutuli D, Cauzzi E, Marino R, Viscomi MT, Petrosini L, Puglisi-Allegra S, Melone M, Keller F, Mercuri NB, Conti F, D'Amelio M. Nilotinib restores memory function by preventing dopaminergic neuron degeneration in a mouse model of Alzheimer's Disease. Prog Neurobiol 2021; 202:102031. [PMID: 33684513 DOI: 10.1016/j.pneurobio.2021.102031] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 02/15/2021] [Accepted: 02/28/2021] [Indexed: 01/18/2023]
Abstract
What happens precociously to the brain destined to develop Alzheimer's Disease (AD) still remains to be elucidated and this is one reason why effective AD treatments are missing. Recent experimental and clinical studies indicate that the degeneration of the dopaminergic (DA) neurons in the Ventral Tegmental Area (VTA) could be one of the first events occurring in AD. However, the causes of the increased vulnerability of DA neurons in AD are missing. Here, we deeply investigate the physiology of DA neurons in the VTA before, at the onset, and after onset of VTA neurodegeneration. We use the Tg2576 mouse model of AD, overexpressing a mutated form of the human APP, to identify molecular targets that can be manipulated pharmacologically. We show that in Tg2576 mice, DA neurons of the VTA at the onset of degeneration undergo slight but functionally relevant changes in their electrophysiological properties and cell morphology. Importantly, these changes are associated with accumulation of autophagosomes, suggestive of a dysfunctional autophagy, and with enhanced activation of c-Abl, a tyrosine kinase previously implicated in the pathogenesis of neurodegenerative diseases. Chronic treatment of Tg2576 mice with Nilotinib, a validated c-Abl inhibitor, reduces c-Abl phosphorylation, improves autophagy, reduces Aβ levels and - more importantly - prevents degeneration as well as functional and morphological alterations in DA neurons of the VTA. Interestingly, the drug prevents the reduction of DA outflow to the hippocampus and ameliorates hippocampal-related cognitive functions. Our results strive to identify early pathological brain changes in AD, to provide a rational basis for new therapeutic interventions able to slow down the disease progression.
Collapse
Affiliation(s)
- Livia La Barbera
- Department of Medicine and Surgery, Department of Sciences and Technologies for Humans and Environment, University Campus Bio-Medico, 00128, Rome, Italy; Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
| | - Francescangelo Vedele
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy; Department of Systems Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Annalisa Nobili
- Department of Medicine and Surgery, Department of Sciences and Technologies for Humans and Environment, University Campus Bio-Medico, 00128, Rome, Italy; Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
| | - Paraskevi Krashia
- Department of Medicine and Surgery, Department of Sciences and Technologies for Humans and Environment, University Campus Bio-Medico, 00128, Rome, Italy; Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy.
| | - Elena Spoleti
- Department of Medicine and Surgery, Department of Sciences and Technologies for Humans and Environment, University Campus Bio-Medico, 00128, Rome, Italy
| | | | - Debora Cutuli
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy; Department of Psychology, Sapienza University of Rome, 00185, Rome, Italy
| | - Emma Cauzzi
- Department of Medicine and Surgery, Department of Sciences and Technologies for Humans and Environment, University Campus Bio-Medico, 00128, Rome, Italy; School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Ramona Marino
- Department of Medicine and Surgery, Department of Sciences and Technologies for Humans and Environment, University Campus Bio-Medico, 00128, Rome, Italy
| | - Maria Teresa Viscomi
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy; Department of Life Science and Public Health Section of Histology and Embryology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Laura Petrosini
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
| | | | - Marcello Melone
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche (UNIVPM), 60020, Ancona, Italy; Center for Neurobiology of Aging, IRCCS Istituto Nazionale Ricovero e Cura Anziani (INRCA), 60020, Ancona, Italy
| | - Flavio Keller
- Department of Medicine and Surgery, Department of Sciences and Technologies for Humans and Environment, University Campus Bio-Medico, 00128, Rome, Italy
| | - Nicola Biagio Mercuri
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy; Department of Systems Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Fiorenzo Conti
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche (UNIVPM), 60020, Ancona, Italy; Center for Neurobiology of Aging, IRCCS Istituto Nazionale Ricovero e Cura Anziani (INRCA), 60020, Ancona, Italy; Foundation for Molecular Medicine, Università Politecnica delle Marche, 60020, Ancona, Italy
| | - Marcello D'Amelio
- Department of Medicine and Surgery, Department of Sciences and Technologies for Humans and Environment, University Campus Bio-Medico, 00128, Rome, Italy; Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy.
| |
Collapse
|
24
|
Nihira H, Izawa K, Ito M, Umebayashi H, Okano T, Kajikawa S, Nanishi E, Keino D, Murakami K, Isa-Nishitani M, Shiba T, Honda Y, Hijikata A, Yasu T, Kubota T, Hasegawa Y, Kawashima Y, Nakano N, Takada H, Ohga S, Heike T, Takita J, Ohara O, Takei S, Takahashi M, Kanegane H, Morio T, Iwaki-Egawa S, Sasahara Y, Nishikomori R, Yasumi T. Detailed analysis of Japanese patients with adenosine deaminase 2 deficiency reveals characteristic elevation of type II interferon signature and STAT1 hyperactivation. J Allergy Clin Immunol 2021; 148:550-562. [PMID: 33529688 DOI: 10.1016/j.jaci.2021.01.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 01/15/2021] [Accepted: 01/21/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Deficiency of adenosine deaminase 2 (DADA2) is an autosomal recessive inflammatory disease caused by loss-of-function mutations in both alleles of the ADA2 gene. Most patients with DADA2 exhibit systemic vasculopathy consistent with polyarteritis nodosa, but large phenotypic variability has been reported, and the pathogenesis of DADA2 remains unclear. OBJECTIVES This study sought to assess the clinical and genetic characteristics of Japanese patients with DADA2 and to gain insight into the pathogenesis of DADA2 by multi-omics analysis. METHODS Clinical and genetic data were collected from 8 Japanese patients with DADA2 diagnosed between 2016 and 2019. ADA2 variants in this cohort were functionally analyzed by in vitro overexpression analysis. PBMCs from 4 patients with DADA2 were subjected to transcriptome and proteome analyses. Patient samples were collected before and after introduction of anti- TNF-α therapies. Transcriptome data were compared with those of normal controls and patients with other autoinflammatory diseases. RESULTS Five novel ADA2 variants were identified in these 8 patients and were confirmed pathogenic by in vitro analysis. Anti-TNF-α therapy controlled inflammation in all 8 patients. Transcriptome and proteome analyses showed that upregulation of type II interferon signaling was characteristic of DADA2. Network analysis identified STAT1 as a key regulator and a hub molecule in DADA2 pathogenesis, a finding supported by the hyperactivation of STAT1 in patients' monocytes and B cells after IFN-γ stimulation. CONCLUSIONS Type II interferon signaling and STAT1 are associated with the pathogenesis of DADA2.
Collapse
Affiliation(s)
- Hiroshi Nihira
- Department of Pediatrics, Kyoto University, Kyoto, Japan
| | - Kazushi Izawa
- Department of Pediatrics, Kyoto University, Kyoto, Japan.
| | - Moeko Ito
- Department of Pharmacy, Hokkaido University of Science, Sapporo, Japan
| | | | - Tsubasa Okano
- Department of Pediatrics and Development Biology, Tokyo Medical and Dental University, Tokyo, Japan
| | | | - Etsuro Nanishi
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Dai Keino
- Division of Hematology/Oncology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Kosaku Murakami
- Department of Rheumatology and Clinical Immunology, Kyoto University, Kyoto, Japan
| | | | - Takeshi Shiba
- Department of Pediatrics, Tenri Hospital, Tenri, Japan
| | | | - Atsushi Hijikata
- Department of Bioscience, Nagahama Institute of Bio-Science and Technology, Nagahama, Japan
| | - Tadateru Yasu
- Department of Pediatrics, Nagasaki Medical Center, Omura, Japan
| | - Tomohiro Kubota
- Department of Pediatrics, Kagoshima University, Kagoshima, Japan
| | - Yoshinori Hasegawa
- Department of Applied Genomics, Kazusa DNA Research Institute, Kisarazu, Japan
| | - Yusuke Kawashima
- Department of Applied Genomics, Kazusa DNA Research Institute, Kisarazu, Japan
| | - Naoko Nakano
- Department of Pediatrics, Ehime University, Toon, Japan
| | - Hidetoshi Takada
- Department of Child Health, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toshio Heike
- Department of Pediatrics, Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Japan
| | - Junko Takita
- Department of Pediatrics, Kyoto University, Kyoto, Japan
| | - Osamu Ohara
- Department of Applied Genomics, Kazusa DNA Research Institute, Kisarazu, Japan
| | - Syuji Takei
- Department of Pediatrics, Kagoshima University, Kagoshima, Japan
| | - Makio Takahashi
- Department of Neurology, Osaka Red Cross Hospital, Osaka, Japan
| | - Hirokazu Kanegane
- Department of Child Health and Development, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomohiro Morio
- Department of Pediatrics and Development Biology, Tokyo Medical and Dental University, Tokyo, Japan
| | | | - Yoji Sasahara
- Department of Pediatrics, Tohoku University, Sendai, Japan
| | - Ryuta Nishikomori
- Department of Pediatrics and Child Health, Kurume University School of Medicine, Kurume, Japan
| | | |
Collapse
|
25
|
Feyder M, Plewnia C, Lieberman OJ, Spigolon G, Piccin A, Urbina L, Dehay B, Li Q, Nilsson P, Altun M, Santini E, Sulzer D, Bezard E, Borgkvist A, Fisone G. Involvement of Autophagy in Levodopa-Induced Dyskinesia. Mov Disord 2021; 36:1137-1146. [PMID: 33460487 PMCID: PMC8248404 DOI: 10.1002/mds.28480] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 12/25/2022] Open
Abstract
Background Autophagy is intensively studied in cancer, metabolic and neurodegenerative diseases, but little is known about its role in pathological conditions linked to altered neurotransmission. We examined the involvement of autophagy in levodopa (l‐dopa)‐induced dyskinesia, a frequent motor complication developed in response to standard dopamine replacement therapy in parkinsonian patients. Methods We used mouse and non‐human primate models of Parkinson's disease to examine changes in autophagy associated with chronic l‐dopa administration and to establish a causative link between impaired autophagy and dyskinesia. Results We found that l‐dopa‐induced dyskinesia is associated with accumulation of the autophagy‐specific substrate p62, a marker of autophagy deficiency. Increased p62 was observed in a subset of projection neurons located in the striatum and depended on l‐dopa‐mediated activation of dopamine D1 receptors, and mammalian target of rapamycin. Inhibition of mammalian target of rapamycin complex 1 with rapamycin counteracted the impairment of autophagy produced by l‐dopa, and reduced dyskinesia. The anti‐dyskinetic effect of rapamycin was lost when autophagy was constitutively suppressed in D1 receptor‐expressing striatal neurons, through inactivation of the autophagy‐related gene protein 7. Conclusions These findings indicate that augmented responsiveness at D1 receptors leads to dysregulated autophagy, and results in the emergence of l‐dopa‐induced dyskinesia. They further suggest the enhancement of autophagy as a therapeutic strategy against dyskinesia. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society
Collapse
Affiliation(s)
- Michael Feyder
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Carina Plewnia
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Ori J Lieberman
- Departments of Neurology, Pharmacology and Psychiatry, Columbia University, and New York State Psychiatric Institute, New York, New York, USA
| | - Giada Spigolon
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Alessandro Piccin
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Lidia Urbina
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Benjamin Dehay
- Univ. Bordeaux, CNRS, IMN, UMR 5293, Bordeaux, F-33000, France
| | - Qin Li
- Motac Neuroscience Ltd, Manchester, United Kingdom.,Institute of Laboratory Animal Sciences & China Academy of Medical Sciences, Beijing, China
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Mikael Altun
- Science for Life Laboratory, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Emanuela Santini
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Departments of Neurology, Pharmacology and Psychiatry, Columbia University, and New York State Psychiatric Institute, New York, New York, USA
| | - David Sulzer
- Departments of Neurology, Pharmacology and Psychiatry, Columbia University, and New York State Psychiatric Institute, New York, New York, USA
| | - Erwan Bezard
- Univ. Bordeaux, CNRS, IMN, UMR 5293, Bordeaux, F-33000, France.,Motac Neuroscience Ltd, Manchester, United Kingdom
| | - Anders Borgkvist
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Departments of Neurology, Pharmacology and Psychiatry, Columbia University, and New York State Psychiatric Institute, New York, New York, USA
| | - Gilberto Fisone
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
26
|
FOXP1 negatively regulates intrinsic excitability in D2 striatal projection neurons by promoting inwardly rectifying and leak potassium currents. Mol Psychiatry 2021; 26:1761-1774. [PMID: 33402705 PMCID: PMC8255328 DOI: 10.1038/s41380-020-00995-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 11/18/2020] [Accepted: 12/09/2020] [Indexed: 01/13/2023]
Abstract
Heterozygous loss-of-function mutations in the transcription factor FOXP1 are strongly associated with autism. Dopamine receptor 2 expressing (D2) striatal projection neurons (SPNs) in heterozygous Foxp1 (Foxp1+/-) mice have higher intrinsic excitability. To understand the mechanisms underlying this alteration, we examined SPNs with cell-type specific homozygous Foxp1 deletion to study cell-autonomous regulation by Foxp1. As in Foxp1+/- mice, D2 SPNs had increased intrinsic excitability with homozygous Foxp1 deletion. This effect involved postnatal mechanisms. The hyperexcitability was mainly due to down-regulation of two classes of potassium currents: inwardly rectifying (KIR) and leak (KLeak). Single-cell RNA sequencing data from D2 SPNs with Foxp1 deletion indicated the down-regulation of transcripts of candidate ion channels that may underlie these currents: Kcnj2 and Kcnj4 for KIR and Kcnk2 for KLeak. This Foxp1-dependent regulation was neuron-type specific since these same currents and transcripts were either unchanged, or very little changed, in D1 SPNs with cell-specific Foxp1 deletion. Our data are consistent with a model where FOXP1 negatively regulates the excitability of D2 SPNs through KIR and KLeak by transcriptionally activating their corresponding transcripts. This, in turn, provides a novel example of how a transcription factor may regulate multiple genes to impact neuronal electrophysiological function that depends on the integration of multiple current types - and do this in a cell-specific fashion. Our findings provide initial clues to altered neuronal function and possible therapeutic strategies not only for FOXP1-associated autism but also for other autism forms associated with transcription factor dysfunction.
Collapse
|
27
|
Neuronal Autophagy Regulates Presynaptic Neurotransmission by Controlling the Axonal Endoplasmic Reticulum. Neuron 2020; 109:299-313.e9. [PMID: 33157003 PMCID: PMC7837115 DOI: 10.1016/j.neuron.2020.10.005] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 08/22/2020] [Accepted: 10/05/2020] [Indexed: 12/21/2022]
Abstract
Neurons are known to rely on autophagy for removal of defective proteins or organelles to maintain synaptic neurotransmission and counteract neurodegeneration. In spite of its importance for neuronal health, the physiological substrates of neuronal autophagy in the absence of proteotoxic challenge have remained largely elusive. We use knockout mice conditionally lacking the essential autophagy protein ATG5 and quantitative proteomics to demonstrate that loss of neuronal autophagy causes selective accumulation of tubular endoplasmic reticulum (ER) in axons, resulting in increased excitatory neurotransmission and compromised postnatal viability in vivo. The gain in excitatory neurotransmission is shown to be a consequence of elevated calcium release from ER stores via ryanodine receptors accumulated in axons and at presynaptic sites. We propose a model where neuronal autophagy controls axonal ER calcium stores to regulate neurotransmission in healthy neurons and in the brain. Neuronal autophagy controls the endoplasmic reticulum (ER) in axons Loss of neuronal autophagy leads to increased excitatory neurotransmission Increased neurotransmission is due to elevated calcium release from ER stores
Collapse
|
28
|
Overhoff M, De Bruyckere E, Kononenko NL. Mechanisms of neuronal survival safeguarded by endocytosis and autophagy. J Neurochem 2020; 157:263-296. [PMID: 32964462 DOI: 10.1111/jnc.15194] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/21/2020] [Accepted: 09/08/2020] [Indexed: 12/11/2022]
Abstract
Multiple aspects of neuronal physiology crucially depend on two cellular pathways, autophagy and endocytosis. During endocytosis, extracellular components either unbound or recognized by membrane-localized receptors (termed "cargo") become internalized into plasma membrane-derived vesicles. These can serve to either recycle the material back to the plasma membrane or send it for degradation to lysosomes. Autophagy also uses lysosomes as a terminal degradation point, although instead of degrading the plasma membrane-derived cargo, autophagy eliminates detrimental cytosolic material and intracellular organelles, which are transported to lysosomes by means of double-membrane vesicles, referred to as autophagosomes. Neurons, like all non-neuronal cells, capitalize on autophagy and endocytosis to communicate with the environment and maintain protein and organelle homeostasis. Additionally, the highly polarized, post-mitotic nature of neurons made them adopt these two pathways for cell-specific functions. These include the maintenance of the synaptic vesicle pool in the pre-synaptic terminal and the long-distance transport of signaling molecules. Originally discovered independently from each other, it is now clear that autophagy and endocytosis are closely interconnected and share several common participating molecules. Considering the crucial role of autophagy and endocytosis in cell type-specific functions in neurons, it is not surprising that defects in both pathways have been linked to the pathology of numerous neurodegenerative diseases. In this review, we highlight the recent knowledge of the role of endocytosis and autophagy in neurons with a special focus on synaptic physiology and discuss how impairments in genes coding for autophagy and endocytosis proteins can cause neurodegeneration.
Collapse
Affiliation(s)
- Melina Overhoff
- CECAD Cluster of Excellence, Institute for Genetics, University of Cologne, Cologne, Germany
| | - Elodie De Bruyckere
- CECAD Cluster of Excellence, Institute for Genetics, University of Cologne, Cologne, Germany
| | - Natalia L Kononenko
- CECAD Cluster of Excellence, Institute for Genetics, University of Cologne, Cologne, Germany
| |
Collapse
|
29
|
Stacchiotti A, Corsetti G. Natural Compounds and Autophagy: Allies Against Neurodegeneration. Front Cell Dev Biol 2020; 8:555409. [PMID: 33072744 PMCID: PMC7536349 DOI: 10.3389/fcell.2020.555409] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 09/01/2020] [Indexed: 12/12/2022] Open
Abstract
Prolonging the healthy life span and limiting neurological illness are imperative goals in gerontology. Age-related neurodegeneration is progressive and leads to severe diseases affecting motility, memory, cognitive function, and social life. To date, no effective treatments are available for neurodegeneration and irreversible neuronal loss. Bioactive phytochemicals could represent a natural alternative to ensure active aging and slow onset of neurodegenerative diseases in elderly patients. Autophagy or macroautophagy is an evolutionarily conserved clearing process that is needed to remove aggregate-prone proteins and organelles in neurons and glia. It also is crucial in synaptic plasticity. Aberrant autophagy has a key role in aging and neurodegeneration. Recent evidence indicates that polyphenols like resveratrol and curcumin, flavonoids, like quercetin, polyamine, like spermidine and sugars, like trehalose, limit brain damage in vitro and in vivo. Their common mechanism of action leads to restoration of efficient autophagy by dismantling misfolded proteins and dysfunctional mitochondria. This review focuses on the role of dietary phytochemicals as modulators of autophagy to fight Alzheimer's and Parkinson's diseases, fronto-temporal dementia, amyotrophic lateral sclerosis, and psychiatric disorders. Currently, most studies have involved in vitro or preclinical animal models, and the therapeutic use of phytochemicals in patients remains limited.
Collapse
Affiliation(s)
- Alessandra Stacchiotti
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Interdepartmental University Center of Research "Adaptation and Regeneration of Tissues and Organs (ARTO)," University of Brescia, Brescia, Italy
| | - Giovanni Corsetti
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
30
|
Lieberman OJ, Cartocci V, Pigulevskiy I, Molinari M, Carbonell J, Broseta MB, Post MR, Sulzer D, Borgkvist A, Santini E. mTOR Suppresses Macroautophagy During Striatal Postnatal Development and Is Hyperactive in Mouse Models of Autism Spectrum Disorders. Front Cell Neurosci 2020; 14:70. [PMID: 32296308 PMCID: PMC7136750 DOI: 10.3389/fncel.2020.00070] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 03/11/2020] [Indexed: 01/06/2023] Open
Abstract
Macroautophagy (hereafter referred to as autophagy) plays a critical role in neuronal function related to development and degeneration. Here, we investigated whether autophagy is developmentally regulated in the striatum, a brain region implicated in neurodevelopmental disease. We demonstrate that autophagic flux is suppressed during striatal postnatal development, reaching adult levels around postnatal day 28 (P28). We also find that mTOR signaling, a key regulator of autophagy, increases during the same developmental period. We further show that mTOR signaling is responsible for suppressing autophagy, via regulation of Beclin-1 and VPS34 activity. Finally, we discover that autophagy is downregulated during late striatal postnatal development (P28) in mice with in utero exposure to valproic acid (VPA), an established mouse model of autism spectrum disorder (ASD). VPA-exposed mice also display deficits in striatal neurotransmission and social behavior. Correction of hyperactive mTOR signaling in VPA-exposed mice restores social behavior. These results demonstrate that neurons coopt metabolic signaling cascades to developmentally regulate autophagy and provide additional evidence that mTOR-dependent signaling pathways represent pathogenic signaling cascades in ASD mouse models that are active during specific postnatal windows.
Collapse
Affiliation(s)
- Ori J. Lieberman
- Division of Molecular Therapeutics, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
- Division of Movement Disorders, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | | | - Irena Pigulevskiy
- Division of Movement Disorders, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Maya Molinari
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Josep Carbonell
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | | | - Michael R. Post
- Division of Molecular Therapeutics, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - David Sulzer
- Division of Molecular Therapeutics, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Anders Borgkvist
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Emanuela Santini
- Division of Movement Disorders, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
31
|
Abstract
Macroautophagy/autophagy is implicated in the maintenance of normal neuronal activity through the regulation of synaptic function and plasticity. However, differences in autophagic degradation within different classes of neurons have not been examined. We have recently demonstrated that autophagy plays very different roles in the two closely related principal neurons of the striatum - the spiny projection neurons of the direct (dSPN) and indirect (iSPN) pathways. Behavioral and electrophysiological experiments revealed that the absence of autophagy in either of these SPN pathways produces unique effects on motor learning, dendritic length, and intrinsic excitability. Specifically, autophagy is required for the normal development of synaptic inputs onto dSPNs, while being required for intrinsic excitability in iSPNs. In iSPNs, this occurs through the regulation of the activity of the KCNJ/Kir2 ion channel, and provides a first demonstration of autophagic control of neuronal intrinsic excitability. ABBREVIATIONS ASD: autism spectrum disorders; dSPNs: direct pathway spiny projection neurons; iSPNs: indirect pathway spiny projection neurons; Kir2: inwardly rectifying potassium channel 2.
Collapse
Affiliation(s)
- Irena Pigulevskiy
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons , New York, NY, USA
| | - Ori J Lieberman
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons , New York, NY, USA
| | - David Sulzer
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons , New York, NY, USA.,Department of Pharmacology, Columbia University Vagelos College of Physicians and Surgeons , New York, NY, USA.,Division of Molecular Therapeutics, New York State Psychiatric Institute , New York, NY, USA
| |
Collapse
|
32
|
Lieberman OJ, Cartocci V, Pigulevskiy I, Molinari M, Carbonell J, Broseta MB, Post MR, Sulzer D, Borgkvist A, Santini E. mTOR Suppresses Macroautophagy During Striatal Postnatal Development and Is Hyperactive in Mouse Models of Autism Spectrum Disorders. Front Cell Neurosci 2020; 14:70. [PMID: 32296308 DOI: 10.3389/fncel.2020.00070/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 03/11/2020] [Indexed: 05/20/2023] Open
Abstract
Macroautophagy (hereafter referred to as autophagy) plays a critical role in neuronal function related to development and degeneration. Here, we investigated whether autophagy is developmentally regulated in the striatum, a brain region implicated in neurodevelopmental disease. We demonstrate that autophagic flux is suppressed during striatal postnatal development, reaching adult levels around postnatal day 28 (P28). We also find that mTOR signaling, a key regulator of autophagy, increases during the same developmental period. We further show that mTOR signaling is responsible for suppressing autophagy, via regulation of Beclin-1 and VPS34 activity. Finally, we discover that autophagy is downregulated during late striatal postnatal development (P28) in mice with in utero exposure to valproic acid (VPA), an established mouse model of autism spectrum disorder (ASD). VPA-exposed mice also display deficits in striatal neurotransmission and social behavior. Correction of hyperactive mTOR signaling in VPA-exposed mice restores social behavior. These results demonstrate that neurons coopt metabolic signaling cascades to developmentally regulate autophagy and provide additional evidence that mTOR-dependent signaling pathways represent pathogenic signaling cascades in ASD mouse models that are active during specific postnatal windows.
Collapse
Affiliation(s)
- Ori J Lieberman
- Division of Molecular Therapeutics, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
- Division of Movement Disorders, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | | | - Irena Pigulevskiy
- Division of Movement Disorders, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Maya Molinari
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Josep Carbonell
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | | | - Michael R Post
- Division of Molecular Therapeutics, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - David Sulzer
- Division of Molecular Therapeutics, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Anders Borgkvist
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Emanuela Santini
- Division of Movement Disorders, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|