1
|
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
|
2
|
Rivagorda M, Romeo-Guitart D, Blanchet V, Mailliet F, Boitez V, Barry N, Milunov D, Siopi E, Goudin N, Moriceau S, Guerrera C, Leibovici M, Saha S, Codogno P, Morselli E, Morel E, Armand AS, Oury F. A primary cilia-autophagy axis in hippocampal neurons is essential to maintain cognitive resilience. NATURE AGING 2025; 5:450-467. [PMID: 39984747 PMCID: PMC11922775 DOI: 10.1038/s43587-024-00791-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 12/06/2024] [Indexed: 02/23/2025]
Abstract
Blood-borne factors are essential to maintain neuronal synaptic plasticity and cognitive resilience throughout life. One such factor is osteocalcin (OCN), a hormone produced by osteoblasts that influences multiple physiological processes, including hippocampal neuronal homeostasis. However, the mechanism through which this blood-borne factor communicates with neurons remains unclear. Here we show the importance of a core primary cilium (PC) protein-autophagy axis in mediating the effects of OCN. We found that the OCN receptor GPR158 is present at the PC of hippocampal neurons and mediates the regulation of autophagy machinery by OCN. During aging, autophagy and PC core proteins are reduced in neurons, and restoring their levels is sufficient to improve cognitive impairments in aged mice. Mechanistically, the induction of this axis by OCN is dependent on the PC-dependent cAMP response element-binding protein signaling pathway. Altogether, this study demonstrates that the PC-autophagy axis is a gateway to mediate communication between blood-borne factors and neurons, and it advances understanding of the mechanisms involved in age-related cognitive decline.
Collapse
Affiliation(s)
- Manon Rivagorda
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - David Romeo-Guitart
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | - Victoria Blanchet
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | - François Mailliet
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | - Valérie Boitez
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | - Natalie Barry
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | | | - Eleni Siopi
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | - Nicolas Goudin
- Platform for Image Analyses, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR 3633, Paris, France
| | - Stéphanie Moriceau
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
- Platform for Neurobehavioral and Metaboblism, Institut Imagine, Structure Fédérative de Recherche Necker, 26 INSERM US24/CNRS UAR, Paris, France
| | - Chiara Guerrera
- Platform for Proteomic Analyses, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR 3633, Paris, France
| | - Michel Leibovici
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | | | - Patrice Codogno
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 6, Paris, France
| | - Eugenia Morselli
- Department of Basic Sciences, Faculty of Medicine and Sciences, Universidad San Sebastián, Santiago, Chile
| | - Etienne Morel
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 6, Paris, France
| | - Anne-Sophie Armand
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | - Franck Oury
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France.
| |
Collapse
|
3
|
Fei L, Liang Y, Kintscher U, Sigrist SJ. Coupling of mitochondrial state with active zone plasticity in early brain aging. Redox Biol 2025; 79:103454. [PMID: 39642596 PMCID: PMC11666929 DOI: 10.1016/j.redox.2024.103454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024] Open
Abstract
Neurodegenerative diseases typically emerge after an extended prodromal period, underscoring the critical importance of initiating interventions during the early stages of brain aging to enhance later resilience. Changes in presynaptic active zone proteins ("PreScale") are considered a dynamic, resilience-enhancing form of plasticity in the process of early, still reversible aging of the Drosophila brain. Aging, however, triggers significant changes not only of synapses but also mitochondria. While the two organelles are spaced in close proximity, likely reflecting a direct functional coupling in regard to ATP and Ca2+ homeostasis, the exact modes of coupling in the aging process remain to understood. We here show that genetic manipulations of mitochondrial functional status, which alters brain oxidative phosphorylation, ATP levels, or the production of reactive oxygen species (ROS), can bidirectionally regulate PreScale during early Drosophila brain aging. Conversely, genetic mimicry of PreScale resulted in decreased oxidative phosphorylation and ATP production, potentially due to reduced mitochondrial calcium (Ca2+) import. Our findings indicate the existence of a positive feedback loop where mitochondrial functional state and PreScale are reciprocally coupled to optimize protection during the early stages of brain aging.
Collapse
Affiliation(s)
- Lu Fei
- Institute for Biology/Genetics, Freie Universität Berlin, 14195, Berlin, Germany
| | - Yongtian Liang
- Institute for Biology/Genetics, Freie Universität Berlin, 14195, Berlin, Germany; NeuroCure Cluster of Excellence, Charité Universitätmedizin Berlin, 10117, Berlin, Germany
| | - Ulrich Kintscher
- Institute of Pharmacology, Center for Cardiovascular Research, Charité Universitätmedizin Berlin, 10115, Berlin, Germany; German Centre for Cardiovascular Research (DZHK), partner site Berlin, 10117, Berlin, Germany
| | - Stephan J Sigrist
- Institute for Biology/Genetics, Freie Universität Berlin, 14195, Berlin, Germany; NeuroCure Cluster of Excellence, Charité Universitätmedizin Berlin, 10117, Berlin, Germany.
| |
Collapse
|
4
|
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
|
5
|
Koch S, Kandimalla P, Padilla E, Kaur S, Kaur R, Nguyen M, Nelson A, Khalsa S, Younossi-Hartenstein A, Hartenstein V. Structural changes shaping the Drosophila ellipsoid body ER-neurons during development and aging. Dev Biol 2024; 516:96-113. [PMID: 39089472 DOI: 10.1016/j.ydbio.2024.07.018] [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: 02/23/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
The ellipsoid body (EB) of the insect brain performs pivotal functions in controlling navigation. Input and output of the EB is provided by multiple classes of R-neurons (now referred to as ER-neurons) and columnar neurons which interact with each other in a stereotypical and spatially highly ordered manner. The developmental mechanisms that control the connectivity and topography of EB neurons are largely unknown. One indispensable prerequisite to unravel these mechanisms is to document in detail the sequence of events that shape EB neurons during their development. In this study, we analyzed the development of the Drosophila EB. In addition to globally following the ER-neuron and columnar neuron (sub)classes in the spatial context of their changing environment we performed a single cell analysis using the multi-color flip out (MCFO) system to analyze the developmental trajectory of ER-neurons at different pupal stages, young adults (4d) and aged adults (∼60d). We show that the EB develops as a merger of two distinct elements, a posterior and anterior EB primordium (prEBp and prEBa, respectively. ER-neurons belonging to different subclasses form growth cones and filopodia that associate with the prEBp and prEBa in a pattern that, from early pupal stages onward, foreshadows their mature structure. Filopodia of all ER-subclasses are initially much longer than the dendritic and terminal axonal branches they give rise to, and are pruned back during late pupal stages. Interestingly, extraneous branches, particularly significant in the dendritic domain, are a hallmark of ER-neuron structure in aged brains. Aging is also associated with a decline in synaptic connectivity from columnar neurons, as well as upregulation of presynaptic protein (Brp) in ER-neurons. Our findings advance the EB (and ER-neurons) as a favorable system to visualize and quantify the development and age-related decline of a complex neuronal circuitry.
Collapse
Affiliation(s)
- Sandra Koch
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Pratyush Kandimalla
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, 90095, USA; Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Eddie Padilla
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Sabrina Kaur
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Rabina Kaur
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - My Nguyen
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Annie Nelson
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Satkartar Khalsa
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Amelia Younossi-Hartenstein
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Volker Hartenstein
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
6
|
Arthur R, Jamwal S, Kumar P. A review on polyamines as promising next-generation neuroprotective and anti-aging therapy. Eur J Pharmacol 2024; 978:176804. [PMID: 38950837 DOI: 10.1016/j.ejphar.2024.176804] [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: 03/25/2024] [Revised: 06/28/2024] [Accepted: 06/28/2024] [Indexed: 07/03/2024]
Abstract
Neurodegenerative disorders are diseases characterized by progressive degeneration of neurons and associated structures and are a major global issue growing more widespread as the global population's average age increases. Despite several investigations on their etiology, the specific cause of these disorders remains unknown. However, there are few symptomatic therapies to treat these disorders. Polyamines (PAs) (putrescine, spermidine, and spermine) are being studied for their role in neuroprotection, aging and cognitive impairment. They are ubiquitous polycations which have relatively higher concentrations in the brain and possess pleiotropic biochemical activities, including regulation of gene expression, ion channels, mitochondria Ca2+ transport, autophagy induction, programmed cell death, and many more. Their cellular content is tightly regulated, and substantial evidence indicates that their altered levels and metabolism are strongly implicated in aging, stress, cognitive dysfunction, and neurodegenerative disorders. In addition, dietary polyamine supplementation has been reported to induce anti-aging effects, anti-oxidant effects, and improve locomotor abnormalities, and cognitive dysfunction. Thus, restoring the polyamine level is considered a promising pharmacological strategy to counteract neurodegeneration. This review highlights PAs' physiological role and the molecular mechanism underpinning their proposed neuroprotective effect in aging and neurodegenerative disorders.
Collapse
Affiliation(s)
- Richmond Arthur
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Sumit Jamwal
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Bathinda, India.
| |
Collapse
|
7
|
Qian M, Zhang N, Zhang R, Liu M, Wu Y, Lu Y, Li F, Zheng L. Non-Linear Association of Dietary Polyamines with the Risk of Incident Dementia: Results from Population-Based Cohort of the UK Biobank. Nutrients 2024; 16:2774. [PMID: 39203912 PMCID: PMC11357304 DOI: 10.3390/nu16162774] [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: 07/17/2024] [Revised: 08/08/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
Natural polyamines, including spermidine (SPD), spermine (SPM) and putrescine (PUT), are evolutionarily conserved endogenous molecules crucially involved in central cellular processes. Their physiological importance may extend to the maintenance of cognitive function during aging. However, limited population-based epidemiological studies have explored the link between dietary polyamines and dementia risk. This study was a prospective analysis of 77,092 UK Biobank participants aged ≥ 60 years without dementia at baseline. We used Cox proportional hazard regression models to explore the associations between dietary polyamines and the risk of dementia, and restricted cubic splines to test the non-linear relationships. During a median follow-up of 12 years, 1087 incidents of all-cause dementia cases occurred, including 450 Alzheimer's disease (AD) cases and 206 vascular dementia (VD) cases. The fully adjusted hazard ratios (HRs) for the upper fourth quintile of dietary SPD, in comparison with the lowest quintile of intake, were 0.68 (95% confidence interval [95% CI]: 0.66-0.83) for the risk of all-cause dementia, 0.62 (95% CI: 0.45-0.85) for AD and 0.56 (95% CI: 0.36-0.88) for VD, respectively. A 26% reduction in dementia risk [HR: 0.74, (95% CI: 0.61-0.89)] and a 47% reduction in AD [HR: 0.53, (95%CI: 0.39-0.72)] were observed comparing the third with the lowest quintiles of dietary SPM. Dietary PUT was only associated with a reduced risk of all-cause dementia in the fourth quintile [HR (95% CI): 0.82 (0.68-0.99)]. Reduced risk was not found to be significant across all quintiles. There were 'U'-shaped relationships found between dietary polyamines and all-cause dementia, AD and VD. Stratification by genetic predisposition showed no significant effect modification. Optimal intake of polyamines was linked to a decreased risk of dementia, with no modification by genetic risk. This potentially suggests cognitive benefits of dietary natural polyamines in humans.
Collapse
Affiliation(s)
- Mingxia Qian
- School of Public Health, Shanghai Jiao Tong University School of Medicine, No. 280 South Chongqing Road, Huangpu District, Shanghai 200025, China; (M.Q.); (N.Z.); (Y.W.)
| | - Na Zhang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, No. 280 South Chongqing Road, Huangpu District, Shanghai 200025, China; (M.Q.); (N.Z.); (Y.W.)
| | - Rui Zhang
- College of Public Health, Shanghai University of Medicine and Health Sciences, No. 279 Zhouzhu Road, Pudong New District, Shanghai 201318, China;
| | - Min Liu
- Department of Epidemiology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang 110122, China;
| | - Yani Wu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, No. 280 South Chongqing Road, Huangpu District, Shanghai 200025, China; (M.Q.); (N.Z.); (Y.W.)
| | - Ying Lu
- Department of Physical and Chemical, Changning District Center for Disease Control and Prevention, Shanghai 200051, China;
| | - Furong Li
- School of Public Health and Emergency Management, Southern University of Science and Technology, No. 1088 Xueyuan Avenue, Nanshan District, Shenzhen 518055, China
| | - Liqiang Zheng
- School of Public Health, Shanghai Jiao Tong University School of Medicine, No. 280 South Chongqing Road, Huangpu District, Shanghai 200025, China; (M.Q.); (N.Z.); (Y.W.)
| |
Collapse
|
8
|
Kakizawa S, Park JJ, Tonoki A. Biology of cognitive aging across species. Geriatr Gerontol Int 2024; 24 Suppl 1:15-24. [PMID: 38126240 DOI: 10.1111/ggi.14782] [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: 08/31/2023] [Revised: 11/27/2023] [Accepted: 12/01/2023] [Indexed: 12/23/2023]
Abstract
Aging is associated with cognitive decline, which can critically affect quality of life. Examining the biology of cognitive aging across species will lead to a better understanding of the fundamental mechanisms involved in this process, and identify potential interventions that could help to improve cognitive function in aging individuals. This minireview aimed to explore the mechanisms and processes involved in cognitive aging across a range of species, from flies to rodents, and covers topics, such as the role of reactive oxygen species and autophagy/mitophagy in cognitive aging. Overall, this literature provides a comprehensive overview of the biology of cognitive aging across species, highlighting the latest research findings and identifying potential avenues for future research. Geriatr Gerontol Int 2024; 24: 15-24.
Collapse
Affiliation(s)
- Sho Kakizawa
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Joong-Jean Park
- Department of Physiology, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Ayako Tonoki
- Department of Biochemistry, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| |
Collapse
|
9
|
Hewson L, Choo A, Webber DL, Trim PJ, Snel MF, Fedele AO, Hopwood JJ, Hemsley KM, O'Keefe LV. Drosophila melanogaster models of MPS IIIC (Hgsnat-deficiency) highlight the role of glia in disease presentation. J Inherit Metab Dis 2024; 47:340-354. [PMID: 38238109 DOI: 10.1002/jimd.12712] [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: 10/03/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 03/16/2024]
Abstract
Sanfilippo syndrome (Mucopolysaccharidosis type III or MPS III) is a recessively inherited neurodegenerative lysosomal storage disorder. Mutations in genes encoding enzymes in the heparan sulphate degradation pathway lead to the accumulation of partially degraded heparan sulphate, resulting ultimately in the development of neurological deficits. Mutations in the gene encoding the membrane protein heparan-α-glucosaminide N-acetyltransferase (HGSNAT; EC2.3.1.78) cause MPS IIIC (OMIM#252930), typified by impaired cognition, sleep-wake cycle changes, hyperactivity and early death, often before adulthood. The precise disease mechanism that causes symptom emergence remains unknown, posing a significant challenge in the development of effective therapeutics. As HGSNAT is conserved in Drosophila melanogaster, we now describe the creation and characterisation of the first Drosophila models of MPS IIIC. Flies with either an endogenous insertion mutation or RNAi-mediated knockdown of hgsnat were confirmed to have a reduced level of HGSNAT transcripts and age-dependent accumulation of heparan sulphate leading to engorgement of the endo/lysosomal compartment. This resulted in abnormalities at the pre-synapse, defective climbing and reduced overall activity. Altered circadian rhythms (shift in peak morning activity) were seen in hgsnat neuronal knockdown lines. Further, when hgsnat was knocked down in specific glial subsets (wrapping, cortical, astrocytes or subperineural glia), impaired climbing or reduced activity was noted, implying that hgsnat function in these specific glial subtypes contributes significantly to this behaviour and targeting treatments to these cell groups may be necessary to ameliorate or prevent symptom onset. These novel models of MPS IIIC provide critical research tools for delineating the key cellular pathways causal in the onset of neurodegeneration in this presently untreatable disorder.
Collapse
Affiliation(s)
- Laura Hewson
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Amanda Choo
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Dani L Webber
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Paul J Trim
- Proteomics, Metabolomics & MS-Imaging Core, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Marten F Snel
- Proteomics, Metabolomics & MS-Imaging Core, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Anthony O Fedele
- Hopwood Centre for Neurobiology, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - John J Hopwood
- Hopwood Centre for Neurobiology, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Kim M Hemsley
- Childhood Dementia Research Group, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Louise V O'Keefe
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
10
|
Wortha SM, Schulz J, Hanna J, Schwarz C, Stubbe B, Frenzel S, Bülow R, Friedrich N, Nauck M, Völzke H, Ewert R, Vogelgesang A, Grabe HJ, Ladenbauer J, Flöel A. Association of spermidine blood levels with microstructure of sleep-implications from a population-based study. GeroScience 2024; 46:1319-1330. [PMID: 37548882 PMCID: PMC10828152 DOI: 10.1007/s11357-023-00886-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/20/2023] [Indexed: 08/08/2023] Open
Abstract
Deteriorations in slow wave sleep (SWS) have been linked to brain aging and Alzheimer's disease (AD), possibly due to its key role in clearance of amyloid-beta and tau (Aß/tau), two pathogenic hallmarks of AD. Spermidine administration has been shown to improve sleep quality in animal models. So far, the association between spermidine levels in humans and parameters of SWS physiology are unknown but may be valuable for therapeutic strategies. Data from 216 participants (age range 50-81 years) of the population-based Study of Health in Pomerania TREND were included in our analysis. We investigated associations between spermidine plasma levels, key parameters of sleep macroarchitecture and microarchitecture that were previously associated with AD pathology, and brain health measured via a marker of structural brain atrophy (AD score). Higher spermidine levels were significantly associated with lower coupling between slow oscillations and spindle activity. No association was evident for SWS, slow oscillatory, and spindle activity throughout non-rapid eye movement sleep. Furthermore, elevated spermidine blood levels were significantly associated with a higher AD score, while sleep markers revealed no association with AD score. The association between higher spermidine levels and brain health was not mediated by coupling between slow oscillations and spindle activity. We report that higher spermidine blood levels are associated not only with deteriorated brain health but also with less advantageous markers of sleep quality in older adults. Future studies need to evaluate whether sleep, spermidine, and Aß/tau deposition are interrelated and whether sleep may play a mediating role.
Collapse
Affiliation(s)
- Silke M Wortha
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany.
- Centre for Mathematical Cognition, School of Science, Loughborough University, Loughborough, UK.
| | - Juliane Schulz
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - Jevri Hanna
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - Claudia Schwarz
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - Beate Stubbe
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Stefan Frenzel
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Robin Bülow
- Institute for Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
| | - Henry Völzke
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Ralf Ewert
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Antje Vogelgesang
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany
| | - Julia Ladenbauer
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - Agnes Flöel
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany
| |
Collapse
|
11
|
Liénard C, Pintart A, Bomont P. Neuronal Autophagy: Regulations and Implications in Health and Disease. Cells 2024; 13:103. [PMID: 38201307 PMCID: PMC10778363 DOI: 10.3390/cells13010103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/02/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
Autophagy is a major degradative pathway that plays a key role in sustaining cell homeostasis, integrity, and physiological functions. Macroautophagy, which ensures the clearance of cytoplasmic components engulfed in a double-membrane autophagosome that fuses with lysosomes, is orchestrated by a complex cascade of events. Autophagy has a particularly strong impact on the nervous system, and mutations in core components cause numerous neurological diseases. We first review the regulation of autophagy, from autophagosome biogenesis to lysosomal degradation and associated neurodevelopmental/neurodegenerative disorders. We then describe how this process is specifically regulated in the axon and in the somatodendritic compartment and how it is altered in diseases. In particular, we present the neuronal specificities of autophagy, with the spatial control of autophagosome biogenesis, the close relationship of maturation with axonal transport, and the regulation by synaptic activity. Finally, we discuss the physiological functions of autophagy in the nervous system, during development and in adulthood.
Collapse
Affiliation(s)
- Caroline Liénard
- NeuroMyoGene Institute—PGNM, CNRS UMR 5261—INSERM U1315, University of Claude Bernard Lyon 1, 69008 Lyon, France; (C.L.); (A.P.)
- CHU Montpellier, University of Montpellier, 34295 Montpellier, France
| | - Alexandre Pintart
- NeuroMyoGene Institute—PGNM, CNRS UMR 5261—INSERM U1315, University of Claude Bernard Lyon 1, 69008 Lyon, France; (C.L.); (A.P.)
| | - Pascale Bomont
- NeuroMyoGene Institute—PGNM, CNRS UMR 5261—INSERM U1315, University of Claude Bernard Lyon 1, 69008 Lyon, France; (C.L.); (A.P.)
| |
Collapse
|
12
|
Chen CC, Lin HW, Feng KL, Tseng DW, de Belle JS, Chiang AS. A subset of cholinergic mushroom body neurons blocks long-term memory formation in Drosophila. Cell Rep 2023; 42:112974. [PMID: 37590142 DOI: 10.1016/j.celrep.2023.112974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 12/22/2022] [Accepted: 07/29/2023] [Indexed: 08/19/2023] Open
Abstract
Long-term memory (LTM) requires learning-induced synthesis of new proteins allocated to specific neurons and synapses in a neural circuit. Not all learned information, however, becomes permanent memory. How the brain gates relevant information into LTM remains unclear. In Drosophila adults, weak learning after a single training session in an olfactory aversive task typically does not induce protein-synthesis-dependent LTM. Instead, strong learning after multiple spaced training sessions is required. Here, we report that pre-synaptic active-zone protein synthesis and cholinergic signaling from the early α/β subset of mushroom body (MB) neurons produce a downstream inhibitory effect on LTM formation. When we eliminated inhibitory signaling from these neurons, weak learning was then sufficient to form LTM. This bidirectional circuit mechanism modulates the transition between distinct memory phase functions in different subpopulations of MB neurons in the olfactory memory circuit.
Collapse
Affiliation(s)
- Chun-Chao Chen
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Hsuan-Wen Lin
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Kuan-Lin Feng
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Der-Wan Tseng
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - J Steven de Belle
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan; School of Life Sciences, Arizona State University, Tempe, AZ 85281, USA; Department of Psychological Sciences, University of San Diego, San Diego, CA 92110, USA; School of Life Sciences, University of Nevada, Las Vegas, NV 89154, USA; MnemOdyssey LLC, Escondido, CA 92027, USA
| | - Ann-Shyn Chiang
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan; Institute of Systems Neuroscience and Department of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan; Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80780, Taiwan; Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli 35053, Taiwan; Graduate Institute of Clinical Medical Science, China Medical University, Taichung 40402, Taiwan; Kavli Institute for Brain and Mind, University of California at San Diego, La Jolla, CA 92093-0526, USA.
| |
Collapse
|
13
|
Ikuta K, Joho D, Kakeyama M, Matsumoto M. Bifidobacterium animalis subsp. lactis and arginine mixture intake improves cognitive flexibility in mice. Front Nutr 2023; 10:1164809. [PMID: 37346910 PMCID: PMC10279864 DOI: 10.3389/fnut.2023.1164809] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/09/2023] [Indexed: 06/23/2023] Open
Abstract
The relationship between intestinal microbiota and cognitive function has been investigated as one of the major topics within the intestinal microbiota-gut-brain axis. Although an increasing number of studies have demonstrated an improvement in learning and memory when using probiotics or prebiotics, to date, there are no studies that target the cognitive flexibility observed in the early stages of several neuropsychiatric diseases, including dementia. We have recently developed a novel behavioral task using the touchscreen operant system to assess cognitive flexibility. We found that the disruption of the intestinal microbiota in mice induced a decline in cognitive flexibility. In the present study, we investigated the effects of treatments consisting of Bifidobacterium animalis subsp. lactis and arginine (Bifal + Arg), which promote the production of intestinal bacterial polyamine, on cognitive flexibility in the mouse model. Male C57BL6 mice orally treated with Bifal + Arg three times a week gradually decreased the 1st-choice incorrect diagonal rate with repeated reversals compared with the control group. Furthermore, in serial reversal phases, Bifal + Arg-treated mice shifted to the behavior of choosing a new correct spot more quickly after the reversal, and this was faster with repeated reversals. These results indicate that this treatment adapts to change and improves cognitive flexibility. This is the first report to show that intestinal environmental control, including probiotics and prebiotics, improves cognitive flexibility in mice.
Collapse
Affiliation(s)
- Kayo Ikuta
- Dairy Science and Technology Institute, Kyodo Milk Industry Co., Ltd., Tokyo, Japan
| | - Daisuke Joho
- Laboratory for Environmental Brain Science, Faculty of Human Sciences, Waseda University, Tokorozawa, Japan
| | - Masaki Kakeyama
- Laboratory for Environmental Brain Science, Faculty of Human Sciences, Waseda University, Tokorozawa, Japan
- Research Institute for Environmental Medical Sciences, Comprehensive Research Organization, Waseda University, Tokorozawa, Japan
| | - Mitsuharu Matsumoto
- Dairy Science and Technology Institute, Kyodo Milk Industry Co., Ltd., Tokyo, Japan
- Research Institute for Environmental Medical Sciences, Comprehensive Research Organization, Waseda University, Tokorozawa, Japan
| |
Collapse
|
14
|
McMullen E, Hertenstein H, Strassburger K, Deharde L, Brankatschk M, Schirmeier S. Glycolytically impaired Drosophila glial cells fuel neural metabolism via β-oxidation. Nat Commun 2023; 14:2996. [PMID: 37225684 DOI: 10.1038/s41467-023-38813-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 05/17/2023] [Indexed: 05/26/2023] Open
Abstract
Neuronal function is highly energy demanding and thus requires efficient and constant metabolite delivery by glia. Drosophila glia are highly glycolytic and provide lactate to fuel neuronal metabolism. Flies are able to survive for several weeks in the absence of glial glycolysis. Here, we study how Drosophila glial cells maintain sufficient nutrient supply to neurons under conditions of impaired glycolysis. We show that glycolytically impaired glia rely on mitochondrial fatty acid breakdown and ketone body production to nourish neurons, suggesting that ketone bodies serve as an alternate neuronal fuel to prevent neurodegeneration. We show that in times of long-term starvation, glial degradation of absorbed fatty acids is essential to ensure survival of the fly. Further, we show that Drosophila glial cells act as a metabolic sensor and can induce mobilization of peripheral lipid stores to preserve brain metabolic homeostasis. Our study gives evidence of the importance of glial fatty acid degradation for brain function, and survival, under adverse conditions in Drosophila.
Collapse
Affiliation(s)
- Ellen McMullen
- Department of Molecular Biology and Genetics, University of South Bohemia, České Budějovice, Czech Republic
| | - Helen Hertenstein
- Zoology and Animal Physiology, Faculty of Biology, Technische Universität Dresden, Dresden, Germany
| | - Katrin Strassburger
- Zoology and Animal Physiology, Faculty of Biology, Technische Universität Dresden, Dresden, Germany
| | - Leon Deharde
- Zoology and Animal Physiology, Faculty of Biology, Technische Universität Dresden, Dresden, Germany
| | - Marko Brankatschk
- Biotechnologisches Zentrum, Technische Universität Dresden, Dresden, Germany.
| | - Stefanie Schirmeier
- Zoology and Animal Physiology, Faculty of Biology, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
15
|
Submerged and Solid-State Fermentation of Spirulina with Lactic Acid Bacteria Strains: Antimicrobial Properties and the Formation of Bioactive Compounds of Protein Origin. BIOLOGY 2023; 12:biology12020248. [PMID: 36829524 PMCID: PMC9952912 DOI: 10.3390/biology12020248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023]
Abstract
The aim of this study was to investigate the changes in bioactive compounds (L-glutamic acid (L-Glu), gamma-aminobutyric acid (GABA) and biogenic amines (BAs)) during the submerged (SMF) and solid-state (SSF) fermentation of Spirulina with lactobacilli strains (Lacticaseibacillus paracasei No. 244; Levilactobacillus brevis No. 173; Leuconostoc mesenteroides No. 225; Liquorilactobacillus uvarum No. 245). The antimicrobial properties of the untreated and fermented Spirulina against a variety of pathogenic and opportunistic strains were tested. The highest concentrations of L-Glu (3841 mg/kg) and GABA (2396 mg/kg) were found after 48 h of SSF with No. 173 and No. 244 strains, respectively. The LAB strain used for biotreatment and the process conditions, as well as the interaction of these factors, had statistically significant effects on the GABA concentration in Spirulina (p ≤ 0.001, p = 0.019 and p = 0.011, respectively). In all cases, the SSF of Spirulina had a higher total BA content than SMF. Most of the fermented Spirulina showed exceptional antimicrobial activity against Staphylococcus aureus but not against the other pathogenic bacteria. The ratios of BA/GABA and BA/L-Glu ranged from 0.5 to 62 and from 0.31 to 10.7, respectively. The GABA content was correlated with putrescine, cadaverine, histamine, tyramine, spermidine and spermine contents. The L-glutamic acid concentration showed positive moderate correlations with tryptamine, putrescine, spermidine and spermine. To summarize, while high concentrations of desirable compounds are formed during fermentation, the formation of non-desirable compounds (BAs) must also be considered due to the similar mechanism of their synthesis as well as the possibility of obtaining high concentrations in the end products.
Collapse
|
16
|
Huang S, Piao C, Beuschel CB, Zhao Z, Sigrist SJ. A brain-wide form of presynaptic active zone plasticity orchestrates resilience to brain aging in Drosophila. PLoS Biol 2022; 20:e3001730. [PMID: 36469518 PMCID: PMC9721493 DOI: 10.1371/journal.pbio.3001730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 11/07/2022] [Indexed: 12/10/2022] Open
Abstract
The brain as a central regulator of stress integration determines what is threatening, stores memories, and regulates physiological adaptations across the aging trajectory. While sleep homeostasis seems to be linked to brain resilience, how age-associated changes intersect to adapt brain resilience to life history remains enigmatic. We here provide evidence that a brain-wide form of presynaptic active zone plasticity ("PreScale"), characterized by increases of active zone scaffold proteins and synaptic vesicle release factors, integrates resilience by coupling sleep, longevity, and memory during early aging of Drosophila. PreScale increased over the brain until mid-age, to then decreased again, and promoted the age-typical adaption of sleep patterns as well as extended longevity, while at the same time it reduced the ability of forming new memories. Genetic induction of PreScale also mimicked early aging-associated adaption of sleep patterns and the neuronal activity/excitability of sleep control neurons. Spermidine supplementation, previously shown to suppress early aging-associated PreScale, also attenuated the age-typical sleep pattern changes. Pharmacological induction of sleep for 2 days in mid-age flies also reset PreScale, restored memory formation, and rejuvenated sleep patterns. Our data suggest that early along the aging trajectory, PreScale acts as an acute, brain-wide form of presynaptic plasticity to steer trade-offs between longevity, sleep, and memory formation in a still plastic phase of early brain aging.
Collapse
Affiliation(s)
- Sheng Huang
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| | - Chengji Piao
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| | - Christine B. Beuschel
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| | - Zhiying Zhao
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - Stephan J. Sigrist
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| |
Collapse
|
17
|
Nasti R, Orlandini S, Furlanetto S, Casale M, Daci A, Hajdari A, Meneghetti F, Villa S, Mori M, Beretta G. An analytical investigation of hydroxylated cinnamoyl polyamines as biomarkers of commercial bee pollen botanical origin. Int J Food Sci Technol 2022. [DOI: http://doi.org/10.1111/ijfs.16142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Rita Nasti
- Department of Environmental Science and Policy (ESP) University of Milan Via G. Celoria 2 20133 Milan Italy
| | - Serena Orlandini
- Department of Chemistry “U. Schiff” University of Florence Via U. Schiff 6, 50019 Sesto Fiorentino Florence Italy
| | - Sandra Furlanetto
- Department of Chemistry “U. Schiff” University of Florence Via U. Schiff 6, 50019 Sesto Fiorentino Florence Italy
| | - Monica Casale
- Department of Pharmacy University of Genova Viale Cembrano, 4 16148 Genova Italy
| | - Armond Daci
- Department of Pharmacy Faculty of Medicine University Hasan Prishtina Pristina Kosovo Albania
| | - Avni Hajdari
- Department of Biology Faculty of Mathematical and Natural Science University of Prishtina Mother Theresa St. 10000 Pristina Kosovo Albania
- Institute of Biological and Environmental Research, University of Prishtina Mother Teresa St. 10000 Pristina Kosovo Albania
| | - Fiorella Meneghetti
- Department of Pharmaceutical Sciences (DISFARM) University of Milan Via L. Mangiagalli 25 20133 Milan Italy
| | - Stefania Villa
- Department of Pharmaceutical Sciences (DISFARM) University of Milan Via L. Mangiagalli 25 20133 Milan Italy
| | - Matteo Mori
- Department of Pharmaceutical Sciences (DISFARM) University of Milan Via L. Mangiagalli 25 20133 Milan Italy
| | - Giangiacomo Beretta
- Department of Environmental Science and Policy (ESP) University of Milan Via G. Celoria 2 20133 Milan Italy
| |
Collapse
|
18
|
Nasti R, Orlandini S, Furlanetto S, Casale M, Daci A, Hajdari A, Meneghetti F, Villa S, Mori M, Beretta G. An analytical investigation of hydroxylated cinnamoyl polyamines as biomarkers of commercial bee pollen botanical origin. Int J Food Sci Technol 2022. [DOI: 10.1111/ijfs.16142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- Rita Nasti
- Department of Environmental Science and Policy (ESP) University of Milan Via G. Celoria 2 20133 Milan Italy
| | - Serena Orlandini
- Department of Chemistry “U. Schiff” University of Florence Via U. Schiff 6, 50019 Sesto Fiorentino Florence Italy
| | - Sandra Furlanetto
- Department of Chemistry “U. Schiff” University of Florence Via U. Schiff 6, 50019 Sesto Fiorentino Florence Italy
| | - Monica Casale
- Department of Pharmacy University of Genova Viale Cembrano, 4 16148 Genova Italy
| | - Armond Daci
- Department of Pharmacy Faculty of Medicine University Hasan Prishtina Pristina Kosovo Albania
| | - Avni Hajdari
- Department of Biology Faculty of Mathematical and Natural Science University of Prishtina Mother Theresa St. 10000 Pristina Kosovo Albania
- Institute of Biological and Environmental Research, University of Prishtina Mother Teresa St. 10000 Pristina Kosovo Albania
| | - Fiorella Meneghetti
- Department of Pharmaceutical Sciences (DISFARM) University of Milan Via L. Mangiagalli 25 20133 Milan Italy
| | - Stefania Villa
- Department of Pharmaceutical Sciences (DISFARM) University of Milan Via L. Mangiagalli 25 20133 Milan Italy
| | - Matteo Mori
- Department of Pharmaceutical Sciences (DISFARM) University of Milan Via L. Mangiagalli 25 20133 Milan Italy
| | - Giangiacomo Beretta
- Department of Environmental Science and Policy (ESP) University of Milan Via G. Celoria 2 20133 Milan Italy
| |
Collapse
|
19
|
Li X, Zhou X, Liu X, Li X, Jiang X, Shi B, Wang S. Spermidine protects against acute kidney injury by modulating macrophage NLRP3 inflammasome activation and mitochondrial respiration in an eIF5A hypusination-related pathway. Mol Med 2022; 28:103. [PMID: 36058905 PMCID: PMC9441050 DOI: 10.1186/s10020-022-00533-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 08/21/2022] [Indexed: 11/26/2022] Open
Abstract
Background Acute kidney injury (AKI) is still a critical problem in clinical practice, with a heavy burden for national health system around the world. It is notable that sepsis is the predominant cause of AKI for patients in the intensive care unit and the mortality remains considerably high. The treatment for AKI relies on supportive therapies and almost no specific treatment is currently available. Spermidine is a naturally occurring polyamine with pleiotropic effects. However, the renoprotective effect of spermidine and the underlying mechanism remain elusive. Methods We employed mice sepsis-induced AKI model and explored the potential renoprotective effect of spermidine in vivo with different administration time and routes. Macrophage depleting was utilized to probe the role of macrophage. In vitro experiments were conducted to examine the effect of spermidine on macrophage cytokine secretion, NLRP3 inflammasome activation and mitochondrial respiration. Results We confirmed that spermidine improves AKI with different administration time and routes and that macrophages serves as an essential mediator in this protective effect. Meanwhile, spermidine downregulates NOD-like receptor protein 3 (NLRP3) inflammasome activation and IL-1 beta production in macrophages directly. Mechanically, spermidine enhances mitochondrial respiration capacity and maintains mitochondria function which contribute to the NLRP3 inhibition. Importantly, we showed that eukaryotic initiation factor 5A (eIF5A) hypusination plays an important role in regulating macrophage bioactivity. Conclusions Spermidine administration practically protects against sepsis-induced AKI in mice and macrophages serve as an essential mediator in this protective effect. Our study identifies spermidine as a promising pharmacologic approach to prevent AKI. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-022-00533-1.
Collapse
Affiliation(s)
- Xianzhi Li
- Department of Urology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China.,Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Jinan, 250014, China
| | - Xiaojun Zhou
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Jinan, 250014, China
| | - Xigao Liu
- Department of Urology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Xiaoyun Li
- Department of Otolaryngology, Qingdao Eighth People's Hospital, Qingdao, 266121, China
| | - Xianzhou Jiang
- Department of Urology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Benkang Shi
- Department of Urology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Shuo Wang
- Department of Urology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China.
| |
Collapse
|
20
|
The Involvement of Polyamines Catabolism in the Crosstalk between Neurons and Astrocytes in Neurodegeneration. Biomedicines 2022; 10:biomedicines10071756. [PMID: 35885061 PMCID: PMC9312548 DOI: 10.3390/biomedicines10071756] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/19/2022] Open
Abstract
In mammalian cells, the content of polyamines is tightly regulated. Polyamines, including spermine, spermidine and putrescine, are involved in many cellular processes. Spermine oxidase specifically oxidizes spermine, and its deregulated activity has been reported to be linked to brain pathologies involving neuron damage. Spermine is a neuromodulator of a number of ionotropic glutamate receptors and types of ion channels. In this respect, the Dach-SMOX mouse model overexpressing spermine oxidase in the neocortex neurons was revealed to be a model of chronic oxidative stress, excitotoxicity and neuronal damage. Reactive astrocytosis, chronic oxidative and excitotoxic stress, neuron loss and the susceptibility to seizure in the Dach-SMOX are discussed here. This genetic model would help researchers understand the linkage between polyamine dysregulation and neurodegeneration and unveil the roles of polyamines in the crosstalk between astrocytes and neurons in neuroprotection or neurodegeneration.
Collapse
|
21
|
Polyamines and Their Metabolism: From the Maintenance of Physiological Homeostasis to the Mediation of Disease. MEDICAL SCIENCES (BASEL, SWITZERLAND) 2022; 10:medsci10030038. [PMID: 35893120 PMCID: PMC9326668 DOI: 10.3390/medsci10030038] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 12/13/2022]
Abstract
The polyamines spermidine and spermine are positively charged aliphatic molecules. They are critical in the regulation of nucleic acid and protein structures, protein synthesis, protein and nucleic acid interactions, oxidative balance, and cell proliferation. Cellular polyamine levels are tightly controlled through their import, export, de novo synthesis, and catabolism. Enzymes and enzymatic cascades involved in polyamine metabolism have been well characterized. This knowledge has been used for the development of novel compounds for research and medical applications. Furthermore, studies have shown that disturbances in polyamine levels and their metabolic pathways, as a result of spontaneous mutations in patients, genetic engineering in mice or experimentally induced injuries in rodents, are associated with multiple maladaptive changes. The adverse effects of altered polyamine metabolism have also been demonstrated in in vitro models. These observations highlight the important role these molecules and their metabolism play in the maintenance of physiological normalcy and the mediation of injury. This review will attempt to cover the extensive and diverse knowledge of the biological role of polyamines and their metabolism in the maintenance of physiological homeostasis and the mediation of tissue injury.
Collapse
|
22
|
Shi YJ, Zhang J, Wang YW, Ding K, Yan Y, Xia CY, Li XX, He J, Zhang WK, Xu JK. The untapped potential of spermidine alkaloids: Sources, structures, bioactivities and syntheses. Eur J Med Chem 2022; 240:114600. [PMID: 35863273 DOI: 10.1016/j.ejmech.2022.114600] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/04/2022] [Accepted: 07/07/2022] [Indexed: 11/26/2022]
Abstract
Spermidine alkaloids are a kind of natural products possessing an aliphatic triamine structure with three or four methylene groups between two N-atoms. Spermidine alkaloids exist in plants, microorganisms, and marine organisms, which usually form amide structures with cinnamic acid or fatty acid derivatives. Their unique structures showed a wide range of biological activities such as neuroprotective, anti-aging, anti-cancer, antioxidant, anti-inflammatory, and antimicrobial. In order to better understand the research status of spermidine alkaloids and promote their applications in human health, this paper systematically reviewed the biological sources, structures, pharmacological actions, and synthetic processes of spermidine alkaloids over the past two decades. This will help to open up new pharmacological investigation fields and better drug design based on these spermidine alkaloids.
Collapse
Affiliation(s)
- Yan-Jing Shi
- School of Life Sciences & School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, PR China; Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Jia Zhang
- School of Life Sciences & School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Yu-Wei Wang
- School of Life Sciences & School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Kang Ding
- School of Life Sciences & School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Yu Yan
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Cong-Yuan Xia
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Xin-Xin Li
- School of Life Sciences & School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Jun He
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China.
| | - Wei-Ku Zhang
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China.
| | - Jie-Kun Xu
- School of Life Sciences & School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, PR China.
| |
Collapse
|
23
|
Kuijpers M. Keeping synapses in shape: degradation pathways in the healthy and aging brain. Neuronal Signal 2022; 6:NS20210063. [PMID: 35813265 PMCID: PMC9208270 DOI: 10.1042/ns20210063] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 11/17/2022] Open
Abstract
Synapses maintain their molecular composition, plasticity and function through the concerted action of protein synthesis and removal. The complex and polarized neuronal architecture poses specific challenges to the logistics of protein and organelle turnover since protein synthesis and degradation mainly happen in the cell soma. In addition, post-mitotic neurons accumulate damage over a lifetime, challenging neuronal degradative pathways and making them particularly susceptible to the effects of aging. This review will summarize the current knowledge on neuronal protein turnover mechanisms with a particular focus on the presynapse, including the proteasome, autophagy and the endolysosomal route and their roles in regulating presynaptic proteostasis and function. In addition, the author will discuss how physiological brain aging, which entails a progressive decline in cognitive functions, affects synapses and the degradative machinery.
Collapse
Affiliation(s)
- Marijn Kuijpers
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Straße 10, 13125 Berlin, Germany
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behaviour and Faculty of Science, Radboud University, Nijmegen, The Netherlands
| |
Collapse
|
24
|
Knodel MM, Dutta Roy R, Wittum G. Influence of T-Bar on Calcium Concentration Impacting Release Probability. Front Comput Neurosci 2022; 16:855746. [PMID: 35586479 PMCID: PMC9108211 DOI: 10.3389/fncom.2022.855746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/09/2022] [Indexed: 11/25/2022] Open
Abstract
The relation of form and function, namely the impact of the synaptic anatomy on calcium dynamics in the presynaptic bouton, is a major challenge of present (computational) neuroscience at a cellular level. The Drosophila larval neuromuscular junction (NMJ) is a simple model system, which allows studying basic effects in a rather simple way. This synapse harbors several special structures. In particular, in opposite to standard vertebrate synapses, the presynaptic boutons are rather large, and they have several presynaptic zones. In these zones, different types of anatomical structures are present. Some of the zones bear a so-called T-bar, a particular anatomical structure. The geometric form of the T-bar resembles the shape of the letter “T” or a table with one leg. When an action potential arises, calcium influx is triggered. The probability of vesicle docking and neurotransmitter release is superlinearly proportional to the concentration of calcium close to the vesicular release site. It is tempting to assume that the T-bar causes some sort of calcium accumulation and hence triggers a higher release probability and thus enhances neurotransmitter exocytosis. In order to study this influence in a quantitative manner, we constructed a typical T-bar geometry and compared the calcium concentration close to the active zones (AZs). We compared the case of synapses with and without T-bars. Indeed, we found a substantial influence of the T-bar structure on the presynaptic calcium concentrations close to the AZs, indicating that this anatomical structure increases vesicle release probability. Therefore, our study reveals how the T-bar zone implies a strong relation between form and function. Our study answers the question of experimental studies (namely “Wichmann and Sigrist, Journal of neurogenetics 2010”) concerning the sense of the anatomical structure of the T-bar.
Collapse
Affiliation(s)
- Markus M. Knodel
- Goethe Center for Scientific Computing (GCSC), Goethe Universität Frankfurt, Frankfurt, Germany
- *Correspondence: Markus M. Knodel ; orcid.org/0000-0001-8739-0803
| | | | - Gabriel Wittum
- Goethe Center for Scientific Computing (GCSC), Goethe Universität Frankfurt, Frankfurt, Germany
- Applied Mathematics and Computational Science, Computer, Electrical and Mathematical Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| |
Collapse
|
25
|
Schwarz C, Benson GS, Horn N, Wurdack K, Grittner U, Schilling R, Märschenz S, Köbe T, Hofer SJ, Magnes C, Stekovic S, Eisenberg T, Sigrist SJ, Schmitz D, Wirth M, Madeo F, Flöel A. Effects of Spermidine Supplementation on Cognition and Biomarkers in Older Adults With Subjective Cognitive Decline: A Randomized Clinical Trial. JAMA Netw Open 2022; 5:e2213875. [PMID: 35616942 PMCID: PMC9136623 DOI: 10.1001/jamanetworkopen.2022.13875] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 03/22/2022] [Indexed: 11/24/2022] Open
Abstract
Importance Developing interventions against age-related memory decline and for older adults experiencing neurodegenerative disease is one of the greatest challenges of our generation. Spermidine supplementation has shown beneficial effects on brain and cognitive health in animal models, and there has been preliminary evidence of memory improvement in individuals with subjective cognitive decline. Objective To determine the effect of longer-term spermidine supplementation on memory performance and biomarkers in this at-risk group. Design, Setting, and Participants This 12-month randomized, double-masked, placebo-controlled phase 2b trial (the SmartAge trial) was conducted between January 2017 and May 2020. The study was a monocenter trial carried out at an academic clinical research center in Germany. Eligible individuals were aged 60 to 90 years with subjective cognitive decline who were recruited from health care facilities as well as through advertisements in the general population. Data analysis was conducted between January and March 2021. Interventions One hundred participants were randomly assigned (1:1 ratio) to 12 months of dietary supplementation with either a spermidine-rich dietary supplement extracted from wheat germ (0.9 mg spermidine/d) or placebo (microcrystalline cellulose). Eighty-nine participants (89%) successfully completed the trial intervention. Main Outcomes and Measures Primary outcome was change in memory performance from baseline to 12-month postintervention assessment (intention-to-treat analysis), operationalized by mnemonic discrimination performance assessed by the Mnemonic Similarity Task. Secondary outcomes included additional neuropsychological, behavioral, and physiological parameters. Safety was assessed in all participants and exploratory per-protocol, as well as subgroup, analyses were performed. Results A total of 100 participants (51 in the spermidine group and 49 in the placebo group) were included in the analysis (mean [SD] age, 69 [5] years; 49 female participants [49%]). Over 12 months, no significant changes were observed in mnemonic discrimination performance (between-group difference, -0.03; 95% CI, -0.11 to 0.05; P = .47) and secondary outcomes. Exploratory analyses indicated possible beneficial effects of the intervention on inflammation and verbal memory. Adverse events were balanced between groups. Conclusions and Relevance In this randomized clinical trial, longer-term spermidine supplementation in participants with subjective cognitive decline did not modify memory and biomarkers compared with placebo. Exploratory analyses indicated possible beneficial effects on verbal memory and inflammation that need to be validated in future studies at higher dosage. Trial Registration ClinicalTrials.gov Identifier: NCT03094546.
Collapse
Affiliation(s)
- Claudia Schwarz
- Department of Neurology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
- NeuroCure Clinical Research Center, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Gloria S. Benson
- Department of Neurology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- NeuroCure Clinical Research Center, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Nora Horn
- Department of Neurology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- NeuroCure Clinical Research Center, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Katharina Wurdack
- Department of Neurology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- NeuroCure Clinical Research Center, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ulrike Grittner
- Institute of Biometry and Clinical Epidemiology, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Ralph Schilling
- Institute of Biometry and Clinical Epidemiology, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
- Institute of Social Medicine, Epidemiology and Health Economics, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Stefanie Märschenz
- NeuroCure Clinical Research Center, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Theresa Köbe
- Department of Neurology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- NeuroCure Clinical Research Center, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases, Dresden, Germany
| | - Sebastian J. Hofer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
| | - Christoph Magnes
- HEALTH–Institute for Biomedicine and Health Sciences, Joanneum Research Forschungsgesellschaft mbH, Graz, Austria
| | - Slaven Stekovic
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Tobias Eisenberg
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
| | - Stephan J. Sigrist
- NeuroCure Clinical Research Center, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - Dietmar Schmitz
- NeuroCure Clinical Research Center, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Miranka Wirth
- Department of Neurology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- NeuroCure Clinical Research Center, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases, Dresden, Germany
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
| | - Agnes Flöel
- German Center for Neurodegenerative Diseases, Greifswald, Germany
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
26
|
Zou D, Zhao Z, Li L, Min Y, Zhang D, Ji A, Jiang C, Wei X, Wu X. A comprehensive review of spermidine: Safety, health effects, absorption and metabolism, food materials evaluation, physical and chemical processing, and bioprocessing. Compr Rev Food Sci Food Saf 2022; 21:2820-2842. [PMID: 35478379 DOI: 10.1111/1541-4337.12963] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/16/2022] [Accepted: 03/21/2022] [Indexed: 12/15/2022]
Abstract
Spermidine, a natural autophagy inducer, has a variety of health effects, such as antitumor, antiaging, anti-inflammation, cardiovascular protection, and neuromodulation. It has been a hot topic in the field of food processing, and current research findings suggest that spermidine-rich foods may be used in intervention and prevention of age-related diseases. In this article, recent findings on the safety, health effects, absorption and metabolism of spermidine were reviewed, and advances in food processing, including the raw materials evaluation, physical and chemical processing, and biological processing of spermidine, were highlighted. In particular, the core metabolic pathways, key gene targets, and efficient metabolic engineering strategies involved in the biosynthesis of spermidine and its precursors were discussed. Moreover, limitations and future perspectives of spermidine research were proposed. The purpose of this review is to provide new insights on spermidine from its safety to its food processing, which will advance the commercial production and applications of spermidine-rich foods and nutraceuticals.
Collapse
Affiliation(s)
- Dian Zou
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Ziyue Zhao
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Lu Li
- Guangdong Key Laboratory of Agricultural Products Processing, Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Yu Min
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Daiyuan Zhang
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Anying Ji
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Cong Jiang
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xuetuan Wei
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xian Wu
- Department of Kinesiology, Nutrition, and Health, Miami University, Oxford, Ohio, USA
| |
Collapse
|
27
|
Gundelfinger ED, Karpova A, Pielot R, Garner CC, Kreutz MR. Organization of Presynaptic Autophagy-Related Processes. Front Synaptic Neurosci 2022; 14:829354. [PMID: 35368245 PMCID: PMC8968026 DOI: 10.3389/fnsyn.2022.829354] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 01/04/2022] [Indexed: 11/13/2022] Open
Abstract
Brain synapses pose special challenges on the quality control of their protein machineries as they are far away from the neuronal soma, display a high potential for plastic adaptation and have a high energy demand to fulfill their physiological tasks. This applies in particular to the presynaptic part where neurotransmitter is released from synaptic vesicles, which in turn have to be recycled and refilled in a complex membrane trafficking cycle. Pathways to remove outdated and damaged proteins include the ubiquitin-proteasome system acting in the cytoplasm as well as membrane-associated endolysosomal and the autophagy systems. Here we focus on the latter systems and review what is known about the spatial organization of autophagy and endolysomal processes within the presynapse. We provide an inventory of which components of these degradative systems were found to be present in presynaptic boutons and where they might be anchored to the presynaptic apparatus. We identify three presynaptic structures reported to interact with known constituents of membrane-based protein-degradation pathways and therefore may serve as docking stations. These are (i) scaffolding proteins of the cytomatrix at the active zone, such as Bassoon or Clarinet, (ii) the endocytic machinery localized mainly at the peri-active zone, and (iii) synaptic vesicles. Finally, we sketch scenarios, how presynaptic autophagic cargos are tagged and recruited and which cellular mechanisms may govern membrane-associated protein turnover in the presynapse.
Collapse
Affiliation(s)
- Eckart D. Gundelfinger
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
- Institute of Pharmacology and Toxicology, Medical Faculty, Otto von Guericke University, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Anna Karpova
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Rainer Pielot
- Institute of Pharmacology and Toxicology, Medical Faculty, Otto von Guericke University, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Craig C. Garner
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Michael R. Kreutz
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- Center for Molecular Neurobiology (ZMNH), University Hospital Hamburg-Eppendorf, Hamburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| |
Collapse
|
28
|
Grochowska KM, Andres‐Alonso M, Karpova A, Kreutz MR. The needs of a synapse—How local organelles serve synaptic proteostasis. EMBO J 2022; 41:e110057. [PMID: 35285533 PMCID: PMC8982616 DOI: 10.15252/embj.2021110057] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/24/2021] [Accepted: 02/10/2022] [Indexed: 12/12/2022] Open
Abstract
Synaptic function crucially relies on the constant supply and removal of neuronal membranes. The morphological complexity of neurons poses a significant challenge for neuronal protein transport since the machineries for protein synthesis and degradation are mainly localized in the cell soma. In response to this unique challenge, local micro‐secretory systems have evolved that are adapted to the requirements of neuronal membrane protein proteostasis. However, our knowledge of how neuronal proteins are synthesized, trafficked to membranes, and eventually replaced and degraded remains scarce. Here, we review recent insights into membrane trafficking at synaptic sites and into the contribution of local organelles and micro‐secretory pathways to synaptic function. We describe the role of endoplasmic reticulum specializations in neurons, Golgi‐related organelles, and protein complexes like retromer in the synthesis and trafficking of synaptic transmembrane proteins. We discuss the contribution of autophagy and of proteasome‐mediated and endo‐lysosomal degradation to presynaptic proteostasis and synaptic function, as well as nondegradative roles of autophagosomes and lysosomes in signaling and synapse remodeling. We conclude that the complexity of neuronal cyto‐architecture necessitates long‐distance protein transport that combines degradation with signaling functions.
Collapse
Affiliation(s)
- Katarzyna M Grochowska
- Leibniz Group “Dendritic Organelles and Synaptic Function” Center for Molecular Neurobiology ZMNH University Medical Center Hamburg‐Eppendorf Hamburg Germany
- Research Group Neuroplasticity Leibniz Institute for Neurobiology Magdeburg Germany
| | - Maria Andres‐Alonso
- Leibniz Group “Dendritic Organelles and Synaptic Function” Center for Molecular Neurobiology ZMNH University Medical Center Hamburg‐Eppendorf Hamburg Germany
- Research Group Neuroplasticity Leibniz Institute for Neurobiology Magdeburg Germany
| | - Anna Karpova
- Research Group Neuroplasticity Leibniz Institute for Neurobiology Magdeburg Germany
- Center for Behavioral Brain Sciences Otto von Guericke University Magdeburg Germany
| | - Michael R Kreutz
- Leibniz Group “Dendritic Organelles and Synaptic Function” Center for Molecular Neurobiology ZMNH University Medical Center Hamburg‐Eppendorf Hamburg Germany
- Research Group Neuroplasticity Leibniz Institute for Neurobiology Magdeburg Germany
- Center for Behavioral Brain Sciences Otto von Guericke University Magdeburg Germany
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg Germany
| |
Collapse
|
29
|
Marcoli M, Cervetto C, Amato S, Fiorucci C, Maura G, Mariottini P, Cervelli M. Transgenic Mouse Overexpressing Spermine Oxidase in Cerebrocortical Neurons: Astrocyte Dysfunction and Susceptibility to Epileptic Seizures. Biomolecules 2022; 12:204. [PMID: 35204705 PMCID: PMC8961639 DOI: 10.3390/biom12020204] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
Polyamines are organic polycations ubiquitously present in living cells. Polyamines are involved in many cellular processes, and their content in mammalian cells is tightly controlled. Among their function, these molecules modulate the activity of several ion channels. Spermine oxidase, specifically oxidized spermine, is a neuromodulator of several types of ion channel and ionotropic glutamate receptors, and its deregulated activity has been linked to several brain pathologies, including epilepsy. The Dach-SMOX mouse line was generated using a Cre/loxP-based recombination approach to study the complex and critical functions carried out by spermine oxidase and spermine in the mammalian brain. This mouse genetic model overexpresses spermine oxidase in the neocortex and is a chronic model of excitotoxic/oxidative injury and neuron vulnerability to oxidative stress and excitotoxic, since its phenotype revealed to be more susceptible to different acute oxidative insults. In this review, the molecular mechanisms underlined the Dach-SMOX phenotype, linked to reactive astrocytosis, neuron loss, chronic oxidative and excitotoxic stress, and susceptibility to seizures have been discussed in detail. The Dach-SMOX mouse model overexpressing SMOX may help in shedding lights on the susceptibility to epileptic seizures, possibly helping to understand the mechanisms underlying epileptogenesis in vulnerable individuals and contributing to provide new molecular mechanism targets to search for novel antiepileptic drugs.
Collapse
Affiliation(s)
- Manuela Marcoli
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genoa, Italy; (S.A.); (G.M.)
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), Lucio Lazzarino 1, 56122 Pisa, Italy
| | - Chiara Cervetto
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genoa, Italy; (S.A.); (G.M.)
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), Lucio Lazzarino 1, 56122 Pisa, Italy
| | - Sarah Amato
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genoa, Italy; (S.A.); (G.M.)
| | - Cristian Fiorucci
- Department of Science, University of Rome “Roma Tre”, Viale Marconi 446, 00146 Rome, Italy; (C.F.); (P.M.)
| | - Guido Maura
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genoa, Italy; (S.A.); (G.M.)
| | - Paolo Mariottini
- Department of Science, University of Rome “Roma Tre”, Viale Marconi 446, 00146 Rome, Italy; (C.F.); (P.M.)
| | - Manuela Cervelli
- Department of Science, University of Rome “Roma Tre”, Viale Marconi 446, 00146 Rome, Italy; (C.F.); (P.M.)
- Neurodevelopment, Neurogenetics and Molecular Neurobiology Unit, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy
| |
Collapse
|
30
|
Cho Y, Kim G, Park J. Mitochondrial aconitase 1 regulates age-related memory impairment via autophagy/mitophagy-mediated neural plasticity in middle-aged flies. Aging Cell 2021; 20:e13520. [PMID: 34799973 PMCID: PMC8672789 DOI: 10.1111/acel.13520] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/06/2021] [Accepted: 11/03/2021] [Indexed: 01/24/2023] Open
Abstract
Age‐related memory impairment (AMI) occurs in many species, including humans. The underlying mechanisms are not fully understood. In wild‐type Drosophila (w1118), AMI appears in the form of a decrease in learning (3‐min memory) from middle age (30 days after eclosion [DAE]). We performed in vivo, DNA microarray, and behavioral screen studies to identify genes controlling both lifespan and AMI and selected mitochondrial Acon1 (mAcon1). mAcon1 expression in the head of w1118 decreased with age. Neuronal overexpression of mAcon1 extended its lifespan and improved AMI. Neuronal or mushroom body expression of mAcon1 regulated the learning of young (10 DAE) and middle‐aged flies. Interestingly, acetyl‐CoA and citrate levels increased in the heads of middle‐aged and neuronal mAcon1 knockdown flies. Acetyl‐CoA, as a cellular energy sensor, is related to autophagy. Autophagy activity and efficacy determined by the positive and negative changes in the expression levels of Atg8a‐II and p62 were proportional to the expression level of mAcon1. Levels of the presynaptic active zone scaffold protein Bruchpilot were inversely proportional to neuronal mAcon1 levels in the whole brain. Furthermore, mAcon1 overexpression in Kenyon cells induced mitophagy labeled with mt‐Keima and improved learning ability. Both processes were blocked by pink1 knockdown. Taken together, our results imply that the regulation of learning and AMI by mAcon1 occurs via autophagy/mitophagy‐mediated neural plasticity.
Collapse
Affiliation(s)
- Yun‐Ho Cho
- Department of Physiology Korea University College of Medicine Seoul Republic of Korea
| | - Gye‐Hyeong Kim
- Department of Physiology Korea University College of Medicine Seoul Republic of Korea
| | - Joong‐Jean Park
- Department of Physiology Korea University College of Medicine Seoul Republic of Korea
| |
Collapse
|
31
|
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
|
32
|
Huang S, Sigrist SJ. Presynaptic and postsynaptic long-term plasticity in sleep homeostasis. Curr Opin Neurobiol 2021; 69:1-10. [DOI: 10.1016/j.conb.2020.11.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/03/2020] [Accepted: 11/15/2020] [Indexed: 12/25/2022]
|
33
|
Banerjee S, Vernon S, Jiao W, Choi BJ, Ruchti E, Asadzadeh J, Burri O, Stowers RS, McCabe BD. Miniature neurotransmission is required to maintain Drosophila synaptic structures during ageing. Nat Commun 2021; 12:4399. [PMID: 34285221 PMCID: PMC8292383 DOI: 10.1038/s41467-021-24490-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 06/22/2021] [Indexed: 11/27/2022] Open
Abstract
The decline of neuronal synapses is an established feature of ageing accompanied by the diminishment of neuronal function, and in the motor system at least, a reduction of behavioural capacity. Here, we have investigated Drosophila motor neuron synaptic terminals during ageing. We observed cumulative fragmentation of presynaptic structures accompanied by diminishment of both evoked and miniature neurotransmission occurring in tandem with reduced motor ability. Through discrete manipulation of each neurotransmission modality, we find that miniature but not evoked neurotransmission is required to maintain presynaptic architecture and that increasing miniature events can both preserve synaptic structures and prolong motor ability during ageing. Our results establish that miniature neurotransmission, formerly viewed as an epiphenomenon, is necessary for the long-term stability of synaptic connections. Synaptic structures disintegrate and fragment as ageing progresses. Here the authors find that miniature neurotransmission is required to maintain adult motor synapse structures in Drosophila and that increasing miniature events can preserve motor ability during ageing.
Collapse
Affiliation(s)
- Soumya Banerjee
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Samuel Vernon
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Wei Jiao
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Ben Jiwon Choi
- Department of Biology, New York University, New York, USA
| | - Evelyne Ruchti
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Jamshid Asadzadeh
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Olivier Burri
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - R Steven Stowers
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, USA
| | - Brian D McCabe
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland.
| |
Collapse
|
34
|
Limanaqi F, Busceti CL, Celli R, Biagioni F, Fornai F. Autophagy as a gateway for the effects of methamphetamine: From neurotransmitter release and synaptic plasticity to psychiatric and neurodegenerative disorders. Prog Neurobiol 2021; 204:102112. [PMID: 34171442 DOI: 10.1016/j.pneurobio.2021.102112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/27/2021] [Accepted: 06/18/2021] [Indexed: 02/07/2023]
Abstract
As a major eukaryotic cell clearing machinery, autophagy grants cell proteostasis, which is key for neurotransmitter release, synaptic plasticity, and neuronal survival. In line with this, besides neuropathological events, autophagy dysfunctions are bound to synaptic alterations that occur in mental disorders, and early on, in neurodegenerative diseases. This is also the case of methamphetamine (METH) abuse, which leads to psychiatric disturbances and neurotoxicity. While consistently altering the autophagy machinery, METH produces behavioral and neurotoxic effects through molecular and biochemical events that can be recapitulated by autophagy blockade. These consist of altered physiological dopamine (DA) release, abnormal stimulation of DA and glutamate receptors, as well as oxidative, excitotoxic, and neuroinflammatory events. Recent molecular insights suggest that METH early impairs the autophagy machinery, though its functional significance remains to be investigated. Here we discuss evidence suggesting that alterations of DA transmission and autophagy are intermingled within a chain of events underlying behavioral alterations and neurodegenerative phenomena produced by METH. Understanding how METH alters the autophagy machinery is expected to provide novel insights into the neurobiology of METH addiction sharing some features with psychiatric disorders and parkinsonism.
Collapse
Affiliation(s)
- Fiona Limanaqi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma, 55, 56126, Pisa, PI, Italy
| | | | - Roberta Celli
- IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli, IS, Italy
| | | | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma, 55, 56126, Pisa, PI, Italy; IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli, IS, Italy.
| |
Collapse
|
35
|
Nakanishi S, Cleveland JL. Polyamine Homeostasis in Development and Disease. MEDICAL SCIENCES (BASEL, SWITZERLAND) 2021; 9:medsci9020028. [PMID: 34068137 PMCID: PMC8162569 DOI: 10.3390/medsci9020028] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/02/2021] [Accepted: 05/06/2021] [Indexed: 12/12/2022]
Abstract
Polycationic polyamines are present in nearly all living organisms and are essential for mammalian cell growth and survival, and for development. These positively charged molecules are involved in a variety of essential biological processes, yet their underlying mechanisms of action are not fully understood. Several studies have shown both beneficial and detrimental effects of polyamines on human health. In cancer, polyamine metabolism is frequently dysregulated, and elevated polyamines have been shown to promote tumor growth and progression, suggesting that targeting polyamines is an attractive strategy for therapeutic intervention. In contrast, polyamines have also been shown to play critical roles in lifespan, cardiac health and in the development and function of the brain. Accordingly, a detailed understanding of mechanisms that control polyamine homeostasis in human health and disease is needed to develop safe and effective strategies for polyamine-targeted therapy.
Collapse
|
36
|
Liang Y, Piao C, Beuschel CB, Toppe D, Kollipara L, Bogdanow B, Maglione M, Lützkendorf J, See JCK, Huang S, Conrad TOF, Kintscher U, Madeo F, Liu F, Sickmann A, Sigrist SJ. eIF5A hypusination, boosted by dietary spermidine, protects from premature brain aging and mitochondrial dysfunction. Cell Rep 2021; 35:108941. [PMID: 33852845 DOI: 10.1016/j.celrep.2021.108941] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 02/11/2021] [Accepted: 03/12/2021] [Indexed: 01/08/2023] Open
Abstract
Mitochondrial function declines during brain aging and is suspected to play a key role in age-induced cognitive decline and neurodegeneration. Supplementing levels of spermidine, a body-endogenous metabolite, has been shown to promote mitochondrial respiration and delay aspects of brain aging. Spermidine serves as the amino-butyl group donor for the synthesis of hypusine (Nε-[4-amino-2-hydroxybutyl]-lysine) at a specific lysine residue of the eukaryotic translation initiation factor 5A (eIF5A). Here, we show that in the Drosophila brain, hypusinated eIF5A levels decline with age but can be boosted by dietary spermidine. Several genetic regimes of attenuating eIF5A hypusination all similarly affect brain mitochondrial respiration resembling age-typical mitochondrial decay and also provoke a premature aging of locomotion and memory formation in adult Drosophilae. eIF5A hypusination, conserved through all eukaryotes as an obviously critical effector of spermidine, might thus be an important diagnostic and therapeutic avenue in aspects of brain aging provoked by mitochondrial decline.
Collapse
Affiliation(s)
- YongTian Liang
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin 14195, Germany; NeuroCure Cluster of Excellence, Charité Universitätmedizin Berlin, Berlin 10117, Germany
| | - Chengji Piao
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin 14195, Germany; NeuroCure Cluster of Excellence, Charité Universitätmedizin Berlin, Berlin 10117, Germany
| | - Christine B Beuschel
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin 14195, Germany; NeuroCure Cluster of Excellence, Charité Universitätmedizin Berlin, Berlin 10117, Germany
| | - David Toppe
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin 14195, Germany; NeuroCure Cluster of Excellence, Charité Universitätmedizin Berlin, Berlin 10117, Germany
| | - Laxmikanth Kollipara
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund 44139, Germany
| | - Boris Bogdanow
- Department of Chemical Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany
| | - Marta Maglione
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin 14195, Germany; NeuroCure Cluster of Excellence, Charité Universitätmedizin Berlin, Berlin 10117, Germany
| | - Janine Lützkendorf
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin 14195, Germany; NeuroCure Cluster of Excellence, Charité Universitätmedizin Berlin, Berlin 10117, Germany
| | - Jason Chun Kit See
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin 14195, Germany; NeuroCure Cluster of Excellence, Charité Universitätmedizin Berlin, Berlin 10117, Germany
| | - Sheng Huang
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin 14195, Germany; NeuroCure Cluster of Excellence, Charité Universitätmedizin Berlin, Berlin 10117, Germany
| | - Tim O F Conrad
- Institute for Mathematics and Computer Sciences, Freie Universität Berlin, Berlin 14195, Germany; Zuse Institute Berlin, Berlin 14195, Germany
| | - Ulrich Kintscher
- German Centre for Cardiovascular Research (DZHK), partner site Berlin, Berlin 10117, Germany; Institute of Pharmacology, Center for Cardiovascular Research, Charité Universitätmedizin Berlin, Berlin 10115, Germany
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria; BioTechMed Graz, Graz, Austria
| | - Fan Liu
- Department of Chemical Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund 44139, Germany; Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen AB24 3FX, Scotland, UK; Medizinische Fakultät, Medizinische Proteom-Center (MPC), Ruhr-Universität Bochum, Bochum 44801, Germany
| | - Stephan J Sigrist
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin 14195, Germany; NeuroCure Cluster of Excellence, Charité Universitätmedizin Berlin, Berlin 10117, Germany.
| |
Collapse
|
37
|
Schroeder S, Hofer SJ, Zimmermann A, Pechlaner R, Dammbrueck C, Pendl T, Marcello GM, Pogatschnigg V, Bergmann M, Müller M, Gschiel V, Ristic S, Tadic J, Iwata K, Richter G, Farzi A, Üçal M, Schäfer U, Poglitsch M, Royer P, Mekis R, Agreiter M, Tölle RC, Sótonyi P, Willeit J, Mairhofer B, Niederkofler H, Pallhuber I, Rungger G, Tilg H, Defrancesco M, Marksteiner J, Sinner F, Magnes C, Pieber TR, Holzer P, Kroemer G, Carmona-Gutierrez D, Scorrano L, Dengjel J, Madl T, Sedej S, Sigrist SJ, Rácz B, Kiechl S, Eisenberg T, Madeo F. Dietary spermidine improves cognitive function. Cell Rep 2021; 35:108985. [PMID: 33852843 DOI: 10.1016/j.celrep.2021.108985] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 02/08/2021] [Accepted: 03/22/2021] [Indexed: 12/22/2022] Open
Abstract
Decreased cognitive performance is a hallmark of brain aging, but the underlying mechanisms and potential therapeutic avenues remain poorly understood. Recent studies have revealed health-protective and lifespan-extending effects of dietary spermidine, a natural autophagy-promoting polyamine. Here, we show that dietary spermidine passes the blood-brain barrier in mice and increases hippocampal eIF5A hypusination and mitochondrial function. Spermidine feeding in aged mice affects behavior in homecage environment tasks, improves spatial learning, and increases hippocampal respiratory competence. In a Drosophila aging model, spermidine boosts mitochondrial respiratory capacity, an effect that requires the autophagy regulator Atg7 and the mitophagy mediators Parkin and Pink1. Neuron-specific Pink1 knockdown abolishes spermidine-induced improvement of olfactory associative learning. This suggests that the maintenance of mitochondrial and autophagic function is essential for enhanced cognition by spermidine feeding. Finally, we show large-scale prospective data linking higher dietary spermidine intake with a reduced risk for cognitive impairment in humans.
Collapse
Affiliation(s)
- Sabrina Schroeder
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria; BioTechMed-Graz, 8010 Graz, Austria
| | - Sebastian J Hofer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria; BioTechMed-Graz, 8010 Graz, Austria; Field of Excellence BioHealth, University of Graz, 8010 Graz, Austria
| | - Andreas Zimmermann
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria; Field of Excellence BioHealth, University of Graz, 8010 Graz, Austria
| | - Raimund Pechlaner
- Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | | | - Tobias Pendl
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - G Mark Marcello
- Department of Anatomy and Histology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary
| | - Viktoria Pogatschnigg
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Martina Bergmann
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Melanie Müller
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Verena Gschiel
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Selena Ristic
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Jelena Tadic
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria; Field of Excellence BioHealth, University of Graz, 8010 Graz, Austria
| | - Keiko Iwata
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy; Research Center for Child Mental Development, University of Fukui, 910-1193 Fukui, Japan; Department of Biology, University of Padova, 35121 Padova, Italy
| | - Gesa Richter
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging Molecular Biology and Biochemistry Medical University of Graz, 8010 Graz, Austria
| | - Aitak Farzi
- Otto Loewi Research Center (for Vascular Biology, Immunology and Inflammation), Division of Pharmacology, Medical University of Graz (MUG), 8010 Graz, Austria
| | - Muammer Üçal
- Department of Neurosurgery, RU Experimental Neurotraumatology, Medical University Graz, 8036 Graz, Austria
| | - Ute Schäfer
- Department of Neurosurgery, RU Experimental Neurotraumatology, Medical University Graz, 8036 Graz, Austria
| | - Michael Poglitsch
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Philipp Royer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Ronald Mekis
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Marlene Agreiter
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Regine C Tölle
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Péter Sótonyi
- Department of Anatomy and Histology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary
| | - Johann Willeit
- Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | | | | | | | | | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Michaela Defrancesco
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Josef Marksteiner
- Department of Psychiatry and Psychotherapy A, Hall State Hospital, 6060 Hall in Tirol, Austria
| | - Frank Sinner
- HEALTH-Institute for Biomedicine and Health Sciences, Joanneum Research Forschungsgesellschaft mbH, 8010 Graz, Austria; Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Christoph Magnes
- HEALTH-Institute for Biomedicine and Health Sciences, Joanneum Research Forschungsgesellschaft mbH, 8010 Graz, Austria
| | - Thomas R Pieber
- BioTechMed-Graz, 8010 Graz, Austria; HEALTH-Institute for Biomedicine and Health Sciences, Joanneum Research Forschungsgesellschaft mbH, 8010 Graz, Austria; Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Peter Holzer
- Otto Loewi Research Center (for Vascular Biology, Immunology and Inflammation), Division of Pharmacology, Medical University of Graz (MUG), 8010 Graz, Austria
| | - Guido Kroemer
- Equipe Labellisée par la Ligue Contre le Cancer, Université Paris Descartes, Université Paris Diderot, Université Sorbonne Paris Cité, INSERM U1138, Centre de Recherche des Cordeliers, 75006 Paris, France; Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, 94 805 Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France; Suzhou Institute for Systems Biology, Chinese Academy of Sciences, 215123 Suzhou, China; Department of Women's and Children's Health, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | | | - Luca Scorrano
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy; Department of Biology, University of Padova, 35121 Padova, Italy
| | - Jörn Dengjel
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Tobias Madl
- BioTechMed-Graz, 8010 Graz, Austria; Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging Molecular Biology and Biochemistry Medical University of Graz, 8010 Graz, Austria
| | - Simon Sedej
- BioTechMed-Graz, 8010 Graz, Austria; Department of Cardiology, Medical University of Graz, 8036 Graz, Austria; Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
| | - Stephan J Sigrist
- Institute of Biology/Genetics, Freie Universität Berlin, 14195 Berlin, Germany
| | - Bence Rácz
- Department of Anatomy and Histology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary
| | - Stefan Kiechl
- Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria; VASCage, Research Centre on Vascular Ageing and Stroke, 6020 Innsbruck, Austria.
| | - Tobias Eisenberg
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria; BioTechMed-Graz, 8010 Graz, Austria; Field of Excellence BioHealth, University of Graz, 8010 Graz, Austria.
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria; BioTechMed-Graz, 8010 Graz, Austria; Field of Excellence BioHealth, University of Graz, 8010 Graz, Austria.
| |
Collapse
|
38
|
Wirth A, Wolf B, Huang CK, Glage S, Hofer SJ, Bankstahl M, Bär C, Thum T, Kahl KG, Sigrist SJ, Madeo F, Bankstahl JP, Ponimaskin E. Novel aspects of age-protection by spermidine supplementation are associated with preserved telomere length. GeroScience 2021; 43:673-690. [PMID: 33517527 PMCID: PMC8110654 DOI: 10.1007/s11357-020-00310-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 12/02/2020] [Indexed: 12/17/2022] Open
Abstract
Ageing provokes a plethora of molecular, cellular and physiological deteriorations, including heart failure, neurodegeneration, metabolic maladaptation, telomere attrition and hair loss. Interestingly, on the molecular level, the capacity to induce autophagy, a cellular recycling and cleaning process, declines with age across a large spectrum of model organisms and is thought to be responsible for a subset of age-induced changes. Here, we show that a 6-month administration of the natural autophagy inducer spermidine in the drinking water to aged mice is sufficient to significantly attenuate distinct age-associated phenotypes. These include modulation of brain glucose metabolism, suppression of distinct cardiac inflammation parameters, decreased number of pathological sights in kidney and liver and decrease of age-induced hair loss. Interestingly, spermidine-mediated age protection was associated with decreased telomere attrition, arguing in favour of a novel cellular mechanism behind the anti-ageing effects of spermidine administration.
Collapse
Affiliation(s)
- Alexander Wirth
- Cellular Neurophysiology, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Bettina Wolf
- Preclinical Molecular Imaging, Department of Nuclear Medicine, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Cheng-Kai Huang
- Institute of Molecular and Translational Therapeutic Strategies, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Silke Glage
- Institute for Laboratory Animal Science, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Sebastian J Hofer
- Institute of Molecular Biosciences, Karl-Franzens-Universität Graz, Humboldtstraße 50/EG, 8010, Graz, Austria
| | - Marion Bankstahl
- Institute for Laboratory Animal Science, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Christian Bär
- Institute of Molecular and Translational Therapeutic Strategies, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
- REBIRTH Center for Translational Regenerative Medicine, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
- REBIRTH Center for Translational Regenerative Medicine, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Nikolai-Fuchs-Straße 1, 30625, Hannover, Germany
| | - Kai G Kahl
- Dept. of Psychiatry; Social Psychiatry and Psychotherapy, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Stephan J Sigrist
- Freie University Berlin, Institute of Biology, Takusstraße 6, 14195, Berlin, Germany
| | - Frank Madeo
- Institute of Molecular Biosciences, Karl-Franzens-Universität Graz, Humboldtstraße 50/EG, 8010, Graz, Austria
| | - Jens P Bankstahl
- Preclinical Molecular Imaging, Department of Nuclear Medicine, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.
| | - Evgeni Ponimaskin
- Cellular Neurophysiology, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, Gagarin ave. 23, Nizhny Novgorod, Russian Federation, 603950.
| |
Collapse
|
39
|
Pooryasin A, Maglione M, Schubert M, Matkovic-Rachid T, Hasheminasab SM, Pech U, Fiala A, Mielke T, Sigrist SJ. Unc13A and Unc13B contribute to the decoding of distinct sensory information in Drosophila. Nat Commun 2021; 12:1932. [PMID: 33771998 PMCID: PMC7997984 DOI: 10.1038/s41467-021-22180-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 02/26/2021] [Indexed: 12/11/2022] Open
Abstract
The physical distance between presynaptic Ca2+ channels and the Ca2+ sensors triggering the release of neurotransmitter-containing vesicles regulates short-term plasticity (STP). While STP is highly diversified across synapse types, the computational and behavioral relevance of this diversity remains unclear. In the Drosophila brain, at nanoscale level, we can distinguish distinct coupling distances between Ca2+ channels and the (m)unc13 family priming factors, Unc13A and Unc13B. Importantly, coupling distance defines release components with distinct STP characteristics. Here, we show that while Unc13A and Unc13B both contribute to synaptic signalling, they play distinct roles in neural decoding of olfactory information at excitatory projection neuron (ePN) output synapses. Unc13A clusters closer to Ca2+ channels than Unc13B, specifically promoting fast phasic signal transfer. Reduction of Unc13A in ePNs attenuates responses to both aversive and appetitive stimuli, while reduction of Unc13B provokes a general shift towards appetitive values. Collectively, we provide direct genetic evidence that release components of distinct nanoscopic coupling distances differentially control STP to play distinct roles in neural decoding of sensory information. The physical distance between synaptic Ca2+ channels and sensors modulates short-term plasticity (STP). Here, the authors show that synaptic release factors Unc13A and Unc13B distinctly couple with Ca2+ channels and contribute to the neural decoding of distinct sensory information in Drosophila.
Collapse
Affiliation(s)
- Atefeh Pooryasin
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - Marta Maglione
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| | - Marco Schubert
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | | | - Sayed-Mohammad Hasheminasab
- Department of Dermatology, Venereology and Allergology, Charité Universitätsmedizin, Berlin, Germany.,CCU Translational Radiation Oncology, DKTK, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ulrike Pech
- Department of Molecular Neurobiology of Behavior, Johann-Friedrich-Blumenbach-Institute for Zoology and Anthropology, University of Göttingen, Göttingen, Germany.,Laboratory of Neuronal Communication, VIB Center for the Biology of Disease, K.U.Leuven, Leuven, Belgium
| | - André Fiala
- Department of Molecular Neurobiology of Behavior, Johann-Friedrich-Blumenbach-Institute for Zoology and Anthropology, University of Göttingen, Göttingen, Germany
| | - Thorsten Mielke
- Max Planck Institute for Molecular Genetics, Berlin, Microscopy and Cryo-Electron Microscopy Group, Berlin, Germany
| | - Stephan J Sigrist
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany. .,NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany.
| |
Collapse
|
40
|
Koch SC, Nelson A, Hartenstein V. Structural aspects of the aging invertebrate brain. Cell Tissue Res 2021; 383:931-947. [PMID: 33409654 PMCID: PMC7965346 DOI: 10.1007/s00441-020-03314-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/28/2020] [Indexed: 11/26/2022]
Abstract
Aging is characterized by a decline in neuronal function in all animal species investigated so far. Functional changes are accompanied by and may be in part caused by, structurally visible degenerative changes in neurons. In the mammalian brain, normal aging shows abnormalities in dendrites and axons, as well as ultrastructural changes in synapses, rather than global neuron loss. The analysis of the structural features of aging neurons, as well as their causal link to molecular mechanisms on the one hand, and the functional decline on the other hand is crucial in order to understand the aging process in the brain. Invertebrate model organisms like Drosophila and C. elegans offer the opportunity to apply a forward genetic approach to the analysis of aging. In the present review, we aim to summarize findings concerning abnormalities in morphology and ultrastructure in invertebrate brains during normal aging and compare them to what is known for the mammalian brain. It becomes clear that despite of their considerably shorter life span, invertebrates display several age-related changes very similar to the mammalian condition, including the retraction of dendritic and axonal branches at specific locations, changes in synaptic density and increased accumulation of presynaptic protein complexes. We anticipate that continued research efforts in invertebrate systems will significantly contribute to reveal (and possibly manipulate) the molecular/cellular pathways leading to neuronal aging in the mammalian brain.
Collapse
Affiliation(s)
- Sandra C Koch
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles (UCLA), Los Angeles, California, USA
| | - Annie Nelson
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles (UCLA), Los Angeles, California, USA
| | - Volker Hartenstein
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles (UCLA), Los Angeles, California, USA.
| |
Collapse
|
41
|
Autophagy status as a gateway for stress-induced catecholamine interplay in neurodegeneration. Neurosci Biobehav Rev 2021; 123:238-256. [PMID: 33497785 DOI: 10.1016/j.neubiorev.2021.01.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 12/13/2022]
Abstract
The catecholamine-containing brainstem nuclei locus coeruleus (LC) and ventral tegmental area (VTA) are critically involved in stress responses. Alterations of catecholamine systems during chronic stress may contribute to neurodegeneration, including cognitive decline. Stress-related catecholamine alterations, while contributing to anxiety and depression, might accelerate neuronal degeneration by increasing the formation of toxic dopamine and norepinephrine by-products. These, in turn, may impair proteostasis within a variety of cortical and subcortical areas. In particular, the molecular events governing neurotransmission, neuroplasticity, and proteostasis within LC and VTA affect a variety of brain areas. Therefore, we focus on alterations of autophagy machinery in these nuclei as a relevant trigger in this chain of events. In fact, these catecholamine-containing areas are mostly prone to autophagy-dependent neurodegeneration. Thus, we propose a dynamic hypothesis according to which stress-induced autophagy alterations within the LC-VTA network foster a cascade towards early neurodegeneration within these nuclei.
Collapse
|
42
|
Doxaki C, Palikaras K. Neuronal Mitophagy: Friend or Foe? Front Cell Dev Biol 2021; 8:611938. [PMID: 33537304 PMCID: PMC7848077 DOI: 10.3389/fcell.2020.611938] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/22/2020] [Indexed: 12/23/2022] Open
Abstract
Maintenance of neuronal homeostasis is a challenging task, due to unique cellular organization and bioenergetic demands of post-mitotic neurons. It is increasingly appreciated that impairment of mitochondrial homeostasis represents an early sign of neuronal dysfunction that is common in both age-related neurodegenerative as well as in neurodevelopmental disorders. Mitochondrial selective autophagy, known as mitophagy, regulates mitochondrial number ensuring cellular adaptation in response to several intracellular and environmental stimuli. Mounting evidence underlines that deregulation of mitophagy levels has an instructive role in the process of neurodegeneration. Although mitophagy induction mediates the elimination of damaged mitochondria and confers neuroprotection, uncontrolled runaway mitophagy could reduce mitochondrial content overstressing the remaining organelles and eventually triggering neuronal cell death. Unveiling the molecular mechanisms of neuronal mitophagy and its intricate role in neuronal survival and cell death, will assist in the development of novel mitophagy modulators to promote cellular and organismal homeostasis in health and disease.
Collapse
Affiliation(s)
| | - Konstantinos Palikaras
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Crete, Greece
| |
Collapse
|
43
|
Matsumoto M. Prevention of Atherosclerosis by the Induction of Microbial Polyamine Production in the Intestinal Lumen. Biol Pharm Bull 2020; 43:221-229. [PMID: 32009110 DOI: 10.1248/bpb.b19-00855] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Low molecular weight metabolites produced by the intestinal microbiome that have been associated with health and disease as metabolites need to be constantly absorbed from the intestinal lumen and transported to intestinal epithelial cells and blood. Polyamines, especially spermidine and spermine, are bioactive chemicals which promote autophagy and suppress inflammation. The main source of exogenous polyamines is the intestinal lumen, where they are produced by intestinal microbiome. Considering the intestinal microbiome as a manufacturing plant for bioactive substances, we developed a novel hybrid putrescine biosynthesis system strategy, in which the simultaneous intake of Bifidobacterium animalis ssp. lactis LKM512 (Bifal) and arginine (Arg) upregulates the production of the putrescine, a precursor of spermidine and spermine, in the gut by controlling the bacterial metabolism beyond its vast diversity and inter-individual differences. In a clinical trial, healthy individuals with a body mass index near the maximum "healthy" range (25 kg/m3; n = 44) were randomized to consume either normal yogurt containing Bifal and Arg (Bifal + Arg YG) or placebo (normal yogurt) for 12 weeks. The change in reactive hyperemia index determined by EndoPAT from week 0 to 12 in the Bifal + Arg YG group was significantly higher than that in the placebo group, indicating that Bifal + Arg YG intake improved vascular endothelial function. In addition, the concentrations of fecal putrescine and serum spermidine in the Bifal+ Arg YG group were significantly higher than those in the placebo group. These findings suggest that consuming Bifal + Arg YG prevents or reduces atherosclerosis risk by upregulating blood spermidine levels, which subsequently induces autophagy.
Collapse
|
44
|
Zahedi K, Brooks M, Barone S, Rahmati N, Murray Stewart T, Dunworth M, Destefano-Shields C, Dasgupta N, Davidson S, Lindquist DM, Fuller CE, Smith RD, Cleveland JL, Casero RA, Soleimani M. Ablation of polyamine catabolic enzymes provokes Purkinje cell damage, neuroinflammation, and severe ataxia. J Neuroinflammation 2020; 17:301. [PMID: 33054763 PMCID: PMC7559641 DOI: 10.1186/s12974-020-01955-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Polyamine catabolism plays a key role in maintaining intracellular polyamine pools, yet its physiological significance is largely unexplored. Here, we report that the disruption of polyamine catabolism leads to severe cerebellar damage and ataxia, demonstrating the fundamental role of polyamine catabolism in the maintenance of cerebellar function and integrity. METHODS Mice with simultaneous deletion of the two principal polyamine catabolic enzymes, spermine oxidase and spermidine/spermine N1-acetyltransferase (Smox/Sat1-dKO), were generated by the crossbreeding of Smox-KO (Smox-/-) and Sat1-KO (Sat1-/-) animals. Development and progression of tissue injury was monitored using imaging, behavioral, and molecular analyses. RESULTS Smox/Sat1-dKO mice are normal at birth, but develop progressive cerebellar damage and ataxia. The cerebellar injury in Smox/Sat1-dKO mice is associated with Purkinje cell loss and gliosis, leading to neuroinflammation and white matter demyelination during the latter stages of the injury. The onset of tissue damage in Smox/Sat1-dKO mice is not solely dependent on changes in polyamine levels as cerebellar injury was highly selective. RNA-seq analysis and confirmatory studies revealed clear decreases in the expression of Purkinje cell-associated proteins and significant increases in the expression of transglutaminases and markers of neurodegenerative microgliosis and astrocytosis. Further, the α-Synuclein expression, aggregation, and polyamination levels were significantly increased in the cerebellum of Smox/Sat1-dKO mice. Finally, there were clear roles of transglutaminase-2 (TGM2) in the cerebellar pathologies manifest in Smox/Sat1-dKO mice, as pharmacological inhibition of transglutaminases reduced the severity of ataxia and cerebellar injury in Smox/Sat1-dKO mice. CONCLUSIONS These results indicate that the disruption of polyamine catabolism, via coordinated alterations in tissue polyamine levels, elevated transglutaminase activity and increased expression, polyamination, and aggregation of α-Synuclein, leads to severe cerebellar damage and ataxia. These studies indicate that polyamine catabolism is necessary to Purkinje cell survival, and for sustaining the functional integrity of the cerebellum.
Collapse
Affiliation(s)
- Kamyar Zahedi
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
- Research Services, Veterans Affairs Medical Center, Cincinnati, OH, 45220, USA.
- Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA.
- Research Services, Veterans Affairs Medical Center, Albuquerque, NM, 87108, USA.
- Department of Internal Medicine, Division of Nephrology, University of New Mexico College of Medicine, 915 Camino de Salud, Bldg. 289, IDTC 3315, Albuquerque, NM, 87113, USA.
- Present Address: Department of Internal Medicine, Division of Nephrology, University of New Mexico College of Medicine, Albuquerque, NM, 87131, USA.
| | - Marybeth Brooks
- Research Services, Veterans Affairs Medical Center, Cincinnati, OH, 45220, USA
- Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
- Present Address: Department of Internal Medicine, Division of Nephrology, University of New Mexico College of Medicine, Albuquerque, NM, 87131, USA
| | - Sharon Barone
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
- Research Services, Veterans Affairs Medical Center, Cincinnati, OH, 45220, USA
- Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
- Research Services, Veterans Affairs Medical Center, Albuquerque, NM, 87108, USA
- Present Address: Department of Internal Medicine, Division of Nephrology, University of New Mexico College of Medicine, Albuquerque, NM, 87131, USA
| | - Negah Rahmati
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02129, USA
| | - Tracy Murray Stewart
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Matthew Dunworth
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Christina Destefano-Shields
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Nupur Dasgupta
- The Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Steve Davidson
- Department of Anesthesiology and Pain Research Center, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Diana M Lindquist
- Department of Radiology, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Christine E Fuller
- Upstate Medical University Department of Pathology, Syracuse, NY, 13219, USA
| | - Roger D Smith
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - John L Cleveland
- Department of Tumor Biology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Department of Cancer Biology, The Scripps Research Institute, Jupiter, FL, USA
| | - Robert A Casero
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Manoocher Soleimani
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
- Research Services, Veterans Affairs Medical Center, Cincinnati, OH, 45220, USA.
- Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA.
- Research Services, Veterans Affairs Medical Center, Albuquerque, NM, 87108, USA.
- Department of Internal Medicine, Division of Nephrology, University of New Mexico College of Medicine, 915 Camino de Salud, Bldg. 289, IDTC 3315, Albuquerque, NM, 87113, USA.
- Present Address: Department of Internal Medicine, Division of Nephrology, University of New Mexico College of Medicine, Albuquerque, NM, 87131, USA.
| |
Collapse
|
45
|
Proietti E, Rossini S, Grohmann U, Mondanelli G. Polyamines and Kynurenines at the Intersection of Immune Modulation. Trends Immunol 2020; 41:1037-1050. [PMID: 33055013 DOI: 10.1016/j.it.2020.09.007] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022]
Abstract
Polyamines (i.e., putrescine, spermidine, and spermine) are bioactive polycations capable of binding nucleic acids and proteins and modulating signaling pathways. Polyamine functions have been studied most extensively in tumors, where they can promote cell transformation and proliferation. Recently, spermidine was found to exert protective effects in an experimental model of multiple sclerosis (MS) and to confer immunoregulatory properties on dendritic cells (DCs), via the indoleamine 2,3-dioxygenase 1 (IDO1) enzyme. IDO1 converts l-tryptophan into metabolites, collectively known as kynurenines, endowed with several immunoregulatory effects via activation of the arylhydrocarbon receptor (AhR). Because AhR activation increases polyamine production, the emerging scenario has identified polyamines and kynurenines as actors of an immunoregulatory circuitry with potential implications for immunotherapy in autoimmune diseases and cancer.
Collapse
Affiliation(s)
- Elisa Proietti
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Sofia Rossini
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Ursula Grohmann
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| | - Giada Mondanelli
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| |
Collapse
|
46
|
Ghosh I, Sankhe R, Mudgal J, Arora D, Nampoothiri M. Spermidine, an autophagy inducer, as a therapeutic strategy in neurological disorders. Neuropeptides 2020; 83:102083. [PMID: 32873420 DOI: 10.1016/j.npep.2020.102083] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 06/18/2020] [Accepted: 07/26/2020] [Indexed: 02/06/2023]
Abstract
Spermidine is a naturally occurring endogenous polyamine synthesized from diamine putrescine. It is a well-known autophagy inducer that maintains cellular and neuronal homeostasis. Healthy brain development and function are dependent on brain polyamine concentration. Polyamines interact with the opioid system, glutamatergic signaling and neuroinflammation in the neuronal and glial compartments. Among the polyamines, spermidine is found highest in the human brain. Age-linked fluctuations in the spermidine levels may possibly contribute to the impairments in neural network and neurogenesis. Exogenously administered spermidine helps in the treatment of brain diseases. Further, current studies highlight the ability of spermidine to promote longevity by inducing autophagy. Still, the causal neuroprotective mechanism of spermidine in neuronal dysfunction remains unidentified. This review aims to summarize various neuroprotective effects of spermidine related to anti-aging/ anti-inflammatory properties and the prevention of neurotoxicity that helps in achieving beneficial effects in age-related neurological disorder. We also expose the signaling cascades modulated by spermidine which might result in therapeutic action. The present review highlights clinical studies along with in-vivo and in-vitro preclinical studies to provide a new dimension for the therapeutic potential of spermidine in neurological disorders.
Collapse
Affiliation(s)
- Indrani Ghosh
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Runali Sankhe
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Jayesh Mudgal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Devinder Arora
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India; School of Pharmacy and Pharmacology, MHIQ, QUM Network, Griffith University, Gold Coast, Queensland, Australia
| | - Madhavan Nampoothiri
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India.
| |
Collapse
|
47
|
Madeo F, Hofer SJ, Pendl T, Bauer MA, Eisenberg T, Carmona-Gutierrez D, Kroemer G. Nutritional Aspects of Spermidine. Annu Rev Nutr 2020; 40:135-159. [DOI: 10.1146/annurev-nutr-120419-015419] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Natural polyamines (spermidine and spermine) are small, positively charged molecules that are ubiquitously found within organisms and cells. They exert numerous (intra)cellular functions and have been implicated to protect against several age-related diseases. Although polyamine levels decline in a complex age-dependent, tissue-, and cell type–specific manner, they are maintained in healthy nonagenarians and centenarians. Increased polyamine levels, including through enhanced dietary intake, have been consistently linked to improved health and reduced overall mortality. In preclinical models, dietary supplementation with spermidine prolongs life span and health span. In this review, we highlight salient aspects of nutritional polyamine intake and summarize the current knowledge of organismal and cellular uptake and distribution of dietary (and gastrointestinal) polyamines and their impact on human health. We further summarize clinical and epidemiological studies of dietary polyamines.
Collapse
Affiliation(s)
- Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
- Field of Excellence BioHealth, University of Graz, 8010 Graz, Austria
| | - Sebastian J. Hofer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Tobias Pendl
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Maria A. Bauer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Tobias Eisenberg
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
- Field of Excellence BioHealth, University of Graz, 8010 Graz, Austria
- Central Lab Graz Cell Informatics and Analyses (GRACIA), NAWI Graz, University of Graz, 8010 Graz, Austria
| | | | - Guido Kroemer
- Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, F-94805 Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, F-75015 Paris, France
- Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Jiangsu 215163, Suzhou, China
- Department of Women's and Children's Health, Karolinska Institute, Karolinska University, S-17177 Solna, Sweden
| |
Collapse
|
48
|
Activate or Inhibit? Implications of Autophagy Modulation as a Therapeutic Strategy for Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21186739. [PMID: 32937909 PMCID: PMC7554997 DOI: 10.3390/ijms21186739] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/01/2020] [Accepted: 09/04/2020] [Indexed: 12/19/2022] Open
Abstract
Neurodegenerative diseases result in a range of conditions depending on the type of proteinopathy, genes affected or the location of the degeneration in the brain. Proteinopathies such as senile plaques and neurofibrillary tangles in the brain are prominent features of Alzheimer’s disease (AD). Autophagy is a highly regulated mechanism of eliminating dysfunctional organelles and proteins, and plays an important role in removing these pathogenic intracellular protein aggregates, not only in AD, but also in other neurodegenerative diseases. Activating autophagy is gaining interest as a potential therapeutic strategy for chronic diseases featuring protein aggregation and misfolding, including AD. Although autophagy activation is a promising intervention, over-activation of autophagy in neurodegenerative diseases that display impaired lysosomal clearance may accelerate pathology, suggesting that the success of any autophagy-based intervention is dependent on lysosomal clearance being functional. Additionally, the effects of autophagy activation may vary significantly depending on the physiological state of the cell, especially during proteotoxic stress and ageing. Growing evidence seems to favour a strategy of enhancing the efficacy of autophagy by preventing or reversing the impairments of the specific processes that are disrupted. Therefore, it is essential to understand the underlying causes of the autophagy defect in different neurodegenerative diseases to explore possible therapeutic approaches. This review will focus on the role of autophagy during stress and ageing, consequences that are linked to its activation and caveats in modulating this pathway as a treatment.
Collapse
|
49
|
De Risi M, Torromino G, Tufano M, Moriceau S, Pignataro A, Rivagorda M, Carrano N, Middei S, Settembre C, Ammassari‐Teule M, Gardoni F, Mele A, Oury F, De Leonibus E. Mechanisms by which autophagy regulates memory capacity in ageing. Aging Cell 2020; 19:e13189. [PMID: 32729663 PMCID: PMC7511873 DOI: 10.1111/acel.13189] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 06/08/2020] [Accepted: 06/13/2020] [Indexed: 12/25/2022] Open
Abstract
Autophagy agonists have been proposed to slow down neurodegeneration. Spermidine, a polyamine that acts as an autophagy agonist, is currently under clinical trial for the treatment of age‐related memory decline. How Spermidine and other autophagy agonists regulate memory and synaptic plasticity is under investigation. We set up a novel mouse model of mild cognitive impairment (MCI), in which middle‐aged (12‐month‐old) mice exhibit impaired memory capacity, lysosomes engulfed with amyloid fibrils (β‐amyloid and α‐synuclein) and impaired task‐induced GluA1 hippocampal post‐translation modifications. Subchronic treatment with Spermidine as well as the autophagy agonist TAT‐Beclin 1 rescued memory capacity and GluA1 post‐translational modifications by favouring the autophagy/lysosomal‐mediated degradation of amyloid fibrils. These findings provide new mechanistic evidence on the therapeutic relevance of autophagy enhancers which, by improving the degradation of misfolded proteins, slow down age‐related memory decline.
Collapse
Affiliation(s)
- Maria De Risi
- Telethon Institute of Genetics and Medicine Telethon Foundation Pozzuoli Italy
- Institute of Biochemistry and Cell Biology (IBBC) National Research Council Rome Italy
| | - Giulia Torromino
- Telethon Institute of Genetics and Medicine Telethon Foundation Pozzuoli Italy
- Institute of Biochemistry and Cell Biology (IBBC) National Research Council Rome Italy
| | - Michele Tufano
- Telethon Institute of Genetics and Medicine Telethon Foundation Pozzuoli Italy
| | - Stéphanie Moriceau
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1151 Institut Necker Enfants‐Malades (INEM) Université Paris Descartes‐Sorbonne–Paris Cité Paris France
| | - Annabella Pignataro
- Laboratory of Psychobiology Department of Experimental Neurology Santa Lucia Foundation Rome Italy
- Institute of Translational Pharmacology (IFT) National Research Council Rome Italy
| | - Manon Rivagorda
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1151 Institut Necker Enfants‐Malades (INEM) Université Paris Descartes‐Sorbonne–Paris Cité Paris France
| | - Nicolò Carrano
- Department of Pharmacological and Biomolecular Sciences University of Milan Milan Italy
| | - Silvia Middei
- Institute of Biochemistry and Cell Biology (IBBC) National Research Council Rome Italy
| | - Carmine Settembre
- Telethon Institute of Genetics and Medicine Telethon Foundation Pozzuoli Italy
| | - Martine Ammassari‐Teule
- Institute of Biochemistry and Cell Biology (IBBC) National Research Council Rome Italy
- Laboratory of Psychobiology Department of Experimental Neurology Santa Lucia Foundation Rome Italy
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences University of Milan Milan Italy
| | - Andrea Mele
- Department of Biology and Biotechnology "C. Darwin" Sapienza University of Rome Rome Italy
- Center for Research in Neurobiology "D. Bovet" Sapienza University of Rome Rome Italy
| | - Franck Oury
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1151 Institut Necker Enfants‐Malades (INEM) Université Paris Descartes‐Sorbonne–Paris Cité Paris France
| | - Elvira De Leonibus
- Telethon Institute of Genetics and Medicine Telethon Foundation Pozzuoli Italy
- Institute of Biochemistry and Cell Biology (IBBC) National Research Council Rome Italy
| |
Collapse
|
50
|
Autophagy in Neuronal Development and Plasticity. Trends Neurosci 2020; 43:767-779. [PMID: 32800535 DOI: 10.1016/j.tins.2020.07.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 06/03/2020] [Accepted: 07/09/2020] [Indexed: 01/05/2023]
Abstract
Autophagy is a highly conserved intracellular clearance pathway in which cytoplasmic contents are trafficked to the lysosome for degradation. Within neurons, it helps to remove damaged organelles and misfolded or aggregated proteins and has therefore been the subject of intense research in relation to neurodegenerative disease. However, far less is understood about the role of autophagy in other aspects of neuronal physiology. Here we review the literature on the role of autophagy in maintaining neuronal stem cells and in neuronal plasticity in adult life and we discuss how these contribute to structural and functional deficits observed in a range of human disorders.
Collapse
|