1
|
Liu Z, Guo Y, Zhang Y, Gao Y, Ning B. Metabolic reprogramming of astrocytes: Emerging roles of lactate. Neural Regen Res 2026; 21:421-432. [PMID: 39688570 DOI: 10.4103/nrr.nrr-d-24-00776] [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: 07/16/2024] [Accepted: 10/25/2024] [Indexed: 12/18/2024] Open
Abstract
Lactate serves as a key energy metabolite in the central nervous system, facilitating essential brain functions, including energy supply, signaling, and epigenetic modulation. Moreover, it links epigenetic modifications with metabolic reprogramming. Nonetheless, the specific mechanisms and roles of this connection in astrocytes remain unclear. Therefore, this review aims to explore the role and specific mechanisms of lactate in the metabolic reprogramming of astrocytes in the central nervous system. The close relationship between epigenetic modifications and metabolic reprogramming was discussed. Therapeutic strategies for targeting metabolic reprogramming in astrocytes in the central nervous system were also outlined to guide future research in central nervous system diseases. In the nervous system, lactate plays an essential role. However, its mechanism of action as a bridge between metabolic reprogramming and epigenetic modifications in the nervous system requires future investigation. The involvement of lactate in epigenetic modifications is currently a hot research topic, especially in lactylation modification, a key determinant in this process. Lactate also indirectly regulates various epigenetic modifications, such as N6-methyladenosine, acetylation, ubiquitination, and phosphorylation modifications, which are closely linked to several neurological disorders. In addition, exploring the clinical applications and potential therapeutic strategies of lactic acid provides new insights for future neurological disease treatments.
Collapse
Affiliation(s)
- Zeyu Liu
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Yijian Guo
- Department of Spinal Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Ying Zhang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Yulei Gao
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Bin Ning
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
- Department of Spinal Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
2
|
Holden JM, Bossardet OL, Bou Ghanem G, Calkins DJ, Wareham LK. Chronic hyperglycemia alters retinal astrocyte microstructure and uptake of cholera toxin B in a murine model of diabetes. J Neurochem 2025; 169:e16237. [PMID: 39374262 DOI: 10.1111/jnc.16237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/04/2024] [Accepted: 09/24/2024] [Indexed: 10/09/2024]
Abstract
Astrocytes are the principle glial cells of the central nervous system and play an active role in maintaining proper metabolism in surrounding neurons. Because of their involvement in metabolic control, it is likely that their physiology changes in response to metabolic diseases such as diabetes and associated diabetic retinopathy. Here, we investigated whether microstructural changes in astrocyte morphology occur during the early stages of chronic hyperglycemia that may be indicative of early pathogenic programs. We used MORF3 mice in conjunction with streptozotocin-induced hyperglycemia to investigate the morphology of single retinal astrocytes at an early timepoint in diabetic disease. We report that astrocytes initiate a morphological remodeling program, which depends on both the glycemic background and the presence of intravitreal injury, to alter the amount of the neuronal-associated pad and bristle microstructural motifs. Additionally, hyperglycemia increases astrocyte uptake of cholera toxin B, possibly reflecting changes in glycolipid and glycoprotein biosynthesis. Chronic hyperglycemia coupled with intravitreal injection of cholera toxin B also causes extensive leukocyte infiltration into the retina. Our results have important clinical relevance as current therapies for diabetic retinopathy involve intravitreal injection of pharmaceuticals in individuals with often poorly controlled blood glucose levels.
Collapse
Affiliation(s)
- Joseph M Holden
- Department of Ophthalmology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt University, Nashville, Tennessee, USA
| | - Olivia L Bossardet
- Department of Ophthalmology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ghazi Bou Ghanem
- Department of Ophthalmology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - David J Calkins
- Department of Ophthalmology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Lauren K Wareham
- Department of Ophthalmology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
3
|
Xu Z, He S, Begum MM, Han X. Myelin Lipid Alterations in Neurodegenerative Diseases: Landscape and Pathogenic Implications. Antioxid Redox Signal 2024; 41:1073-1099. [PMID: 39575748 PMCID: PMC11971557 DOI: 10.1089/ars.2024.0676] [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: 05/14/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 12/14/2024]
Abstract
Significance: Lipids, which constitute the highest portion (over 50%) of brain dry mass, are crucial for brain integrity, energy homeostasis, and signaling regulation. Emerging evidence revealed that lipid profile alterations and abnormal lipid metabolism occur during normal aging and in different forms of neurodegenerative diseases. Moreover, increasing genome-wide association studies have validated new targets on lipid-associated pathways involved in disease development. Myelin, the protective sheath surrounding axons, is crucial for efficient neural signaling transduction. As the primary site enriched with lipids, impairments of myelin are increasingly recognized as playing significant and complex roles in various neurodegenerative diseases, beyond simply being secondary effects of neuronal loss. Recent Advances: With advances in the lipidomics field, myelin lipid alterations and their roles in contributing to or reflecting the progression of diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, multiple sclerosis, and others, have recently caught great attention. Critical Issues: This review summarizes recent findings of myelin lipid alterations in the five most common neurodegenerative diseases and discusses their implications in disease pathogenesis. Future Directions: By highlighting myelin lipid abnormalities in neurodegenerative diseases, this review aims to encourage further research focused on lipids and the development of new lipid-oriented therapeutic approaches in this area. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Ziying Xu
- Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, Texas, USA
| | - Sijia He
- Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, Texas, USA
| | - Mst Marium Begum
- Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, Texas, USA
| | - Xianlin Han
- Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, Texas, USA
- Department of Medicine, UT Health San Antonio, San Antonio, Texas, USA
| |
Collapse
|
4
|
Müller Y, Lengacher L, Friscourt F, Quairiaux C, Stoppini L, Magistretti PJ, Lengacher S, Finsterwald C. Epileptiform activity in brain organoids derived from patient with Glucose Transporter 1 Deficiency Syndrome. Front Neurosci 2024; 18:1498801. [PMID: 39605786 PMCID: PMC11599213 DOI: 10.3389/fnins.2024.1498801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024] Open
Abstract
Introduction Glucose Transporter 1-Deficiency Syndrome (GLUT1-DS) is a rare genetic disorder caused by mutations in the gene encoding for GLUT1 and characterized by impaired glucose uptake in the brain. This leads to brain hypometabolism and the development of symptoms that include epilepsy, motor dysfunctions and cognitive impairment. The development of patient-specific in vitro models is a valuable tool for understanding the pathophysiology of rare genetic disorders and testing new therapeutic interventions. Methods In this study, we generated brain organoids from induced pluripotent stem cells (iPSCs) derived either from a GLUT1-DS patient or a healthy individual. The functional organoids were analyzed for cellular composition, maturity, and electrophysiological activity using a custom-made microelectrode array (MEA) platform, which allowed for the detection of spikes, burst patterns, and epileptiform discharges. Results Immunostaining revealed a similar distribution of neurons and astrocytes in both healthy and GLUT1-DS brain organoids, though GLUT1-DS brain organoids exhibited reduced cellular density and smaller overall size. Electrophysiological recordings demonstrated functional spike profiles in both organoid types. Notably, our study demonstrates that brain organoids derived from a GLUT1-DS patient exhibit distinct epileptiform activity and heightened sensitivity to glucose deprivation, reflecting key features of the disorder. Discussion These findings validate the use of brain organoids as a model for studying GLUT1-DS and highlight their potential for testing novel therapeutic strategies aimed at improving glucose metabolism and managing epilepsy in patients.
Collapse
Affiliation(s)
| | | | - F. Friscourt
- Functional Brain Mapping Lab, Department of Basic Neuroscience, University of Geneva, Geneva, Switzerland
- Neurosurgery Clinic, Department of Clinical Neuroscience, University Hospital Geneva, Geneva, Switzerland
| | - C. Quairiaux
- Functional Brain Mapping Lab, Department of Basic Neuroscience, University of Geneva, Geneva, Switzerland
| | - L. Stoppini
- Tissue Engineering Laboratory, HEPIA HES-SO University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland
| | | | | | | |
Collapse
|
5
|
Zhong X, D’Antona AM, Rouse JC. Mechanistic and Therapeutic Implications of Protein and Lipid Sialylation in Human Diseases. Int J Mol Sci 2024; 25:11962. [PMID: 39596031 PMCID: PMC11594235 DOI: 10.3390/ijms252211962] [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: 10/08/2024] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Glycan structures of glycoproteins and glycolipids on the surface glycocalyx and luminal sugar layers of intracellular membrane compartments in human cells constitute a key interface between intracellular biological processes and external environments. Sialic acids, a class of alpha-keto acid sugars with a nine-carbon backbone, are frequently found as the terminal residues of these glycoconjugates, forming the critical components of these sugar layers. Changes in the status and content of cellular sialic acids are closely linked to many human diseases such as cancer, cardiovascular, neurological, inflammatory, infectious, and lysosomal storage diseases. The molecular machineries responsible for the biosynthesis of the sialylated glycans, along with their biological interacting partners, are important therapeutic strategies and targets for drug development. The purpose of this article is to comprehensively review the recent literature and provide new scientific insights into the mechanisms and therapeutic implications of sialylation in glycoproteins and glycolipids across various human diseases. Recent advances in the clinical developments of sialic acid-related therapies are also summarized and discussed.
Collapse
Affiliation(s)
- Xiaotian Zhong
- BioMedicine Design, Discovery and Early Development, Pfizer Research and Development, 610 Main Street, Cambridge, MA 02139, USA;
| | - Aaron M. D’Antona
- BioMedicine Design, Discovery and Early Development, Pfizer Research and Development, 610 Main Street, Cambridge, MA 02139, USA;
| | - Jason C. Rouse
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, MA 01810, USA;
| |
Collapse
|
6
|
Li H, Liu Y, Sun Y, Guo H, Lv S, Guo W, Ren J, Wang Y, Zu J, Yan J, Wang N. Targeting astrocytes polarization after spinal cord injury: a promising direction. Front Cell Neurosci 2024; 18:1478741. [PMID: 39479524 PMCID: PMC11521873 DOI: 10.3389/fncel.2024.1478741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024] Open
Abstract
Spinal cord injury (SCI) is a serious neurological injury that causes severe trauma to motor and sensory functions. Although long considered incurable, recent research has brought new hope for functional recovery from SCI. After SCI, astrocytes are activated into many polarization states. Here we discuss the two most important classical phenotypes: the 'A1' neurotoxic phenotype and the 'A2' neuroprotective phenotype, with A1 astrocytes being neurotoxic and impeding neurorecovery, and A2 astrocytes being neuroprotective. This paper discusses the changes in astrocyte responsiveness after SCI and the pros and cons of their polarization in SCI. It also elucidates the feasibility of astrocyte polarization as a therapeutic target for neuroprotection. In the future, multiple intervention strategies targeting astrocyte polarization are expected to gain wider clinical application, ultimately improving motor-sensory function and quality of life in SCI patients.
Collapse
Affiliation(s)
- Helin Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Ying Liu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yucao Sun
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hangyu Guo
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Shiyan Lv
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Wenhui Guo
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jiyu Ren
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yufu Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jianing Zu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jinglong Yan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Nanxiang Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
7
|
d'Avanzo N, Paolino D, Barone A, Ciriolo L, Mancuso A, Christiano MC, Tolomeo AM, Celia C, Deng X, Fresta M. OX26-cojugated gangliosilated liposomes to improve the post-ischemic therapeutic effect of CDP-choline. Drug Deliv Transl Res 2024; 14:2771-2787. [PMID: 38478324 PMCID: PMC11384645 DOI: 10.1007/s13346-024-01556-3] [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] [Accepted: 02/19/2024] [Indexed: 09/10/2024]
Abstract
Cerebrovascular impairment represents one of the main causes of death worldwide with a mortality rate of 5.5 million per year. The disability of 50% of surviving patients has high social impacts and costs in long period treatment for national healthcare systems. For these reasons, the efficacious clinical treatment of patients, with brain ischemic stroke, remains a medical need. To this aim, a liposome nanomedicine, with monosialic ganglioside type 1 (GM1), OX26 (an anti-transferrin receptor antibody), and CDP-choline (a neurotrophic drug) (CDP-choline/OX26Lip) was prepared. CDP-choline/OX26Lip were prepared by a freeze and thaw method and then extruded through polycarbonate filters, to have narrow size distributed liposomes of ~80 nm. CDP-choline/OX26Lip were stable in human serum, they had suitable pharmacokinetic properties, and 30.0 ± 4.2% of the injected drug was still present in the blood stream 12 h after its systemic injection. The post-ischemic therapeutic effect of CDP-choline/OX26Lip is higher than CDP-choline/Lip, thus showing a significantly high survival rate of the re-perfused post-ischemic rats, i.e. 96% and 78% after 8 days. The treatment with CDP-choline/OX26Lip significantly decreased the peroxidation rate of ~5-times compared to CDP-choline/Lip; and the resulting conjugated dienes, that was 13.9 ± 1.1 mmol/mg proteins for CDP-choline/Lip and 3.1 ± 0.8 for CDP-choline/OX26Lip. OX26 increased the accumulation of GM1-liposomes in the brain tissues and thus the efficacious of CDP-choline. Therefore, this nanomedicine may represent a strategy for the reassessment of CDP-choline to treat post-ischemic events caused by brain stroke, and respond to a significant clinical need.
Collapse
Affiliation(s)
- Nicola d'Avanzo
- Department of Clinical and Experimental Medicine, University of Catanzaro "Magna Graecia", Viale "S. Venuta", 88100, Catanzaro, Italy
| | - Donatella Paolino
- Department of Clinical and Experimental Medicine, University of Catanzaro "Magna Graecia", Viale "S. Venuta", 88100, Catanzaro, Italy
| | - Antonella Barone
- Department of Clinical and Experimental Medicine, University of Catanzaro "Magna Graecia", Viale "S. Venuta", 88100, Catanzaro, Italy
| | - Luigi Ciriolo
- Department of Health Sciences, University of Catanzaro "Magna Graecia", Viale "S. Venuta", 88100, Catanzaro, Italy
| | - Antonia Mancuso
- Department of Clinical and Experimental Medicine, University of Catanzaro "Magna Graecia", Viale "S. Venuta", 88100, Catanzaro, Italy
| | - Maria Chiara Christiano
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna Graecia", Viale "S. Venuta", 88100, Catanzaro, Italy
| | - Anna Maria Tolomeo
- Department of Cardiac, Thoracic and Vascular Science and Public Health, University of Padova, 35128, Padua, Italy
- Perdiatric Research Institute "Città della Speranza", Corso Stati Uniti, 4, 35127, Padua, Italy
| | - Christian Celia
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", Via dei Vestini 31, 66100, Chieti, Italy.
- Lithuanian University of Health Sciences, Laboratory of Drug Targets Histopathology, Institute of Cardiology, A. Mickeviciaus g. 9, LT-44307, Kaunas, Lithuania.
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China.
| | - Xiaoyong Deng
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China.
| | - Massimo Fresta
- Department of Health Sciences, University of Catanzaro "Magna Graecia", Viale "S. Venuta", 88100, Catanzaro, Italy.
| |
Collapse
|
8
|
Rha AK, Kan SH, Andrade-Heckman P, Christensen CL, Harb JF, Wang RY. Base editing of the GLB1 gene is therapeutic in GM1 gangliosidosis patient-derived cells. Mol Genet Metab 2024; 143:108568. [PMID: 39303319 DOI: 10.1016/j.ymgme.2024.108568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/19/2024] [Accepted: 08/20/2024] [Indexed: 09/22/2024]
Abstract
GM1 gangliosidosis is an autosomal recessive neurodegenerative lysosomal storage disease caused by pathogenic variants in the GLB1 gene, limiting the production of active lysosomal β-galactosidase. Phenotypic heterogeneity is due in part to variant type, location within GLB1, and the amount of residual enzyme activity; in the most severe form, death occurs in infancy. With no FDA approved therapeutics, development of efficacious strategies for the disease is pivotal. CRISPR/Cas based approaches have revolutionized precision medicine and have been indispensable to the development of treatments for several monogenic disorders with bespoke strategies central to current research pipelines. We used CRISPR/Cas-adenine base editing to correct the GLB1 c.380G>A (p.Cys127Tyr) variant in patient-derived dermal fibroblasts compound heterozygous with the GLB1 c.481T>G (p.Trp161Gly) pathogenic variant. Nucleofection of plasmids encoding the target sgRNA and ABEmax restored the canonical guanine (32.2 ± 2.2 % of the target allele) and synthesis of active β-galactosidase. Analysis of cellular markers of pathology revealed normalization of both primary glycoconjugate storage and lysosomal pathology. Furthermore, analysis of off-target sites nominated by the in silico tools Cas-OFFinder and/or CRISTA revealed no significant editing or indels. This study supports the use of CRISPR/Cas-based approaches for the treatment of GM1 gangliosidosis, and provides foundational data for future translational studies.
Collapse
Affiliation(s)
- Allisandra K Rha
- Research Institute, Children's Hospital of Orange County, Orange, CA 92868, United States
| | - Shih-Hsin Kan
- Research Institute, Children's Hospital of Orange County, Orange, CA 92868, United States
| | - Perla Andrade-Heckman
- Research Institute, Children's Hospital of Orange County, Orange, CA 92868, United States
| | - Chloe L Christensen
- Research Institute, Children's Hospital of Orange County, Orange, CA 92868, United States
| | - Jerry F Harb
- Research Institute, Children's Hospital of Orange County, Orange, CA 92868, United States
| | - Raymond Y Wang
- Division of Metabolic Disorders, Children's Hospital of Orange County Specialists, Orange, CA 92868, United States; Department of Pediatrics, University of California-Irvine School of Medicine, Irvine, CA 92697, United States.
| |
Collapse
|
9
|
Imperlini E, Di Marzio L, Cevenini A, Costanzo M, Nicola d'Avanzo, Fresta M, Orrù S, Celia C, Salvatore F. Unraveling the impact of different liposomal formulations on the plasma protein corona composition might give hints on the targeting capability of nanoparticles. NANOSCALE ADVANCES 2024; 6:4434-4449. [PMID: 39170967 PMCID: PMC11334990 DOI: 10.1039/d4na00345d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 06/27/2024] [Indexed: 08/23/2024]
Abstract
Nanoparticles (NPs) interact with biological fluids after being injected into the bloodstream. The interactions between NPs and plasma proteins at the nano-bio interface affect their biopharmaceutical properties and distribution in the organ and tissues due to protein corona (PrC) composition, and in turn, modification of the resulting targeting capability. Moreover, lipid and polymer NPs, at their interface, affect the composition of PrC and the relative adsorption and abundance of specific proteins. To investigate this latter aspect, we synthesized and characterized different liposomal formulations (LFs) with lipids and polymer-conjugated lipids at different molar ratios, having different sizes, size distributions and surface charges. The PrC composition of various designed LFs was evaluated ex vivo in human plasma by label-free quantitative proteomics. We also correlated the relative abundance of identified specific proteins in the coronas of the different LFs with their physicochemical properties (size, PDI, zeta potential). The evaluation of outputs from different bioinformatic tools discovered protein clusters allowing to highlight: (i) common as well as the unique species for the various formulations; (ii) correlation between each identified PrC and the physicochemical properties of LFs; (iii) some preferential binding determined by physicochemical properties of LFs; (iv) occurrence of formulation-specific protein patterns in PrC. Investigating specific clusters in PrC will help decode the multivalent roles of the protein pattern components in the drug delivery process, taking advantage of the bio-nanoscale recognition and identification for significant advances in nanomedicine.
Collapse
Affiliation(s)
- Esther Imperlini
- Department for Innovation in Biological, Agrofood and Forest Systems, University of Tuscia Viterbo 01100 Italy
| | - Luisa Di Marzio
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio" Via dei Vestini 31 66100 Chieti Italy +39 0871 3554711
| | - Armando Cevenini
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II Naples 80131 Italy +39 3356069177
- CEINGE-Biotecnologie Avanzate Franco Salvatore Naples 80145 Italy +39 081 3737880
| | - Michele Costanzo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II Naples 80131 Italy +39 3356069177
- CEINGE-Biotecnologie Avanzate Franco Salvatore Naples 80145 Italy +39 081 3737880
| | - Nicola d'Avanzo
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Graecia" Viale "S. Venuta" 88100 Catanzaro Italy
- Department of Experimental and Clinical Medicine, Research Center "ProHealth Translational Hub", "Magna Graecia" University of Catanzaro, Campus Universitario "S. Venuta"-Building of BioSciences Viale S. Venuta 88100 Catanzaro Italy
| | - Massimo Fresta
- Department of Experimental and Clinical Medicine, Research Center "ProHealth Translational Hub", "Magna Graecia" University of Catanzaro, Campus Universitario "S. Venuta"-Building of BioSciences Viale S. Venuta 88100 Catanzaro Italy
- Department of Health Sciences, University of Catanzaro "Magna Graecia" Viale "S. Venuta" 88100 Catanzaro Italy
| | - Stefania Orrù
- CEINGE-Biotecnologie Avanzate Franco Salvatore Naples 80145 Italy +39 081 3737880
- Department of Medical, Movement and Wellness Sciences, University of Naples Parthenope Naples 80133 Italy
| | - Christian Celia
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio" Via dei Vestini 31 66100 Chieti Italy +39 0871 3554711
- Lithuanian University of Health Sciences, Laboratory of Drug Targets Histopathology, Institute of Cardiology A. Mickeviciaus g. 9 LT-44307 Kaunas Lithuania
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University Shanghai 200444 China
- UdA-TechLab, Research Center, University of Chieti-Pescara "G. D'Annunzio" 66100 Chieti Italy
| | - Francesco Salvatore
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II Naples 80131 Italy +39 3356069177
- CEINGE-Biotecnologie Avanzate Franco Salvatore Naples 80145 Italy +39 081 3737880
| |
Collapse
|
10
|
Lunghi G, Di Biase E, Carsana EV, Henriques A, Callizot N, Mauri L, Ciampa MG, Mari L, Loberto N, Aureli M, Sonnino S, Spedding M, Chiricozzi E, Fazzari M. GM1 ganglioside exerts protective effects against glutamate-excitotoxicity via its oligosaccharide in wild-type and amyotrophic lateral sclerosis motor neurons. FEBS Open Bio 2023; 13:2324-2341. [PMID: 37885330 PMCID: PMC10699117 DOI: 10.1002/2211-5463.13727] [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: 07/19/2023] [Revised: 10/14/2023] [Accepted: 10/25/2023] [Indexed: 10/28/2023] Open
Abstract
Alterations in glycosphingolipid metabolism have been linked to the pathophysiological mechanisms of amyotrophic lateral sclerosis (ALS), a neurodegenerative disease affecting motor neurons. Accordingly, administration of GM1, a sialic acid-containing glycosphingolipid, is protective against neuronal damage and supports neuronal homeostasis, with these effects mediated by its bioactive component, the oligosaccharide head (GM1-OS). Here, we add new evidence to the therapeutic efficacy of GM1 in ALS: Its administration to WT and SOD1G93A motor neurons affected by glutamate-induced excitotoxicity significantly increased neuronal survival and preserved neurite networks, counteracting intracellular protein accumulation and mitochondria impairment. Importantly, the GM1-OS faithfully replicates GM1 activity, emphasizing that even in ALS the protective function of GM1 strictly depends on its pentasaccharide.
Collapse
Affiliation(s)
- Giulia Lunghi
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanoSegrateItaly
| | - Erika Di Biase
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanoSegrateItaly
| | - Emma Veronica Carsana
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanoSegrateItaly
| | | | | | - Laura Mauri
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanoSegrateItaly
| | - Maria Grazia Ciampa
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanoSegrateItaly
| | - Luigi Mari
- Department of ImmunologySt. Jude Children's Research HospitalMemphisTNUSA
| | - Nicoletta Loberto
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanoSegrateItaly
| | - Massimo Aureli
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanoSegrateItaly
| | - Sandro Sonnino
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanoSegrateItaly
| | | | - Elena Chiricozzi
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanoSegrateItaly
| | - Maria Fazzari
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanoSegrateItaly
| |
Collapse
|
11
|
Orellana AM, Mazucanti CH, Dos Anjos LP, de Sá Lima L, Kawamoto EM, Scavone C. Klotho increases antioxidant defenses in astrocytes and ubiquitin-proteasome activity in neurons. Sci Rep 2023; 13:15080. [PMID: 37699938 PMCID: PMC10497516 DOI: 10.1038/s41598-023-41166-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 08/23/2023] [Indexed: 09/14/2023] Open
Abstract
Klotho is an antiaging protein, and its levels decline with age and chronic stress. The exogenous administration of Klotho can enhance cognitive performance in mice and negatively modulate the Insulin/IGF1/PI3K/AKT pathway in terms of metabolism. In humans, insulin sensitivity is a hallmark of healthy longevity. Therefore, this study aimed to determine if exogenous Klotho, when added to neuronal and astrocytic cell cultures, could reduce the phosphorylation levels of certain insulin signaling effectors and enhance antioxidant strategies in these cells. Primary cell cultures of cortical astrocytes and neurons from mice were exposed to 1 nM Klotho for 24 h, with or without glucose. Klotho decreased pAKT and mTOR levels. However, in astrocytes, Klotho increased FOXO-3a activity and catalase levels, shielding them from intermediate oxidative stress. In neurons, Klotho did not alter FOXO-3 phosphorylation levels but increased proteasome activity, maintaining lower levels of PFKFB3. This study offers new insights into the roles of Klotho in regulating energy metabolism and the redox state in the brain.
Collapse
Affiliation(s)
- Ana Maria Orellana
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences ICB-1, University of São Paulo, Avenida Professor Lineu Prestes, 1524, São Paulo, São Paulo, 05508-900, Brazil
| | - Caio Henrique Mazucanti
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences ICB-1, University of São Paulo, Avenida Professor Lineu Prestes, 1524, São Paulo, São Paulo, 05508-900, Brazil
- Laboratory of Clinical Investigation, Diabetes Section, National Institute on Aging (NIH/NIA), Baltimore, MD, USA
| | - Leticia Pavan Dos Anjos
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences ICB-1, University of São Paulo, Avenida Professor Lineu Prestes, 1524, São Paulo, São Paulo, 05508-900, Brazil
| | - Larissa de Sá Lima
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences ICB-1, University of São Paulo, Avenida Professor Lineu Prestes, 1524, São Paulo, São Paulo, 05508-900, Brazil
| | - Elisa Mitiko Kawamoto
- Laboratory of Molecular and Functional Neurobiology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Cristoforo Scavone
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences ICB-1, University of São Paulo, Avenida Professor Lineu Prestes, 1524, São Paulo, São Paulo, 05508-900, Brazil.
| |
Collapse
|
12
|
Spanos F, Deleidi M. Glycolipids in Parkinson's disease: beyond neuronal function. FEBS Open Bio 2023; 13:1558-1579. [PMID: 37219461 PMCID: PMC10476577 DOI: 10.1002/2211-5463.13651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/10/2023] [Accepted: 05/22/2023] [Indexed: 05/24/2023] Open
Abstract
Glycolipid balance is key to normal body function, and its alteration can lead to a variety of diseases involving multiple organs and tissues. Glycolipid disturbances are also involved in Parkinson's disease (PD) pathogenesis and aging. Increasing evidence suggests that glycolipids affect cellular functions beyond the brain, including the peripheral immune system, intestinal barrier, and immunity. Hence, the interplay between aging, genetic predisposition, and environmental exposures could initiate systemic and local glycolipid changes that lead to inflammatory reactions and neuronal dysfunction. In this review, we discuss recent advances in the link between glycolipid metabolism and immune function and how these metabolic changes can exacerbate immunological contributions to neurodegenerative diseases, with a focus on PD. Further understanding of the cellular and molecular mechanisms that control glycolipid pathways and their impact on both peripheral tissues and the brain will help unravel how glycolipids shape immune and nervous system communication and the development of novel drugs to prevent PD and promote healthy aging.
Collapse
Affiliation(s)
- Fokion Spanos
- Institut Imagine, INSERM UMR1163Paris Cité UniversityFrance
- Aligning Science Across Parkinson's (ASAP) Collaborative Research NetworkChevy ChaseMDUSA
| | - Michela Deleidi
- Institut Imagine, INSERM UMR1163Paris Cité UniversityFrance
- Aligning Science Across Parkinson's (ASAP) Collaborative Research NetworkChevy ChaseMDUSA
- Department of Neurodegenerative Diseases, Center of Neurology, Hertie Institute for Clinical Brain ResearchUniversity of TübingenGermany
| |
Collapse
|
13
|
Schneider JS. GM1 Ganglioside as a Disease-Modifying Therapeutic for Parkinson's Disease: A Multi-Functional Glycosphingolipid That Targets Multiple Parkinson's Disease-Relevant Pathogenic Mechanisms. Int J Mol Sci 2023; 24:9183. [PMID: 37298133 PMCID: PMC10252733 DOI: 10.3390/ijms24119183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder affecting millions of patients worldwide. Many therapeutics are available for treating PD symptoms but there is no disease-modifying therapeutic that has been unequivocally shown to slow or stop the progression of the disease. There are several factors contributing to the failure of many putative disease-modifying agents in clinical trials and these include the choice of patients and clinical trial designs for disease modification trials. Perhaps more important, however, is the choice of therapeutic, which for the most part, has not taken into account the multiple and complex pathogenic mechanisms and processes involved in PD. This paper discusses some of the factors contributing to the lack of success in PD disease-modification trials, which have mostly investigated therapeutics with a singular mechanism of action directed at one of the many PD pathogenic processes, and suggests that an alternative strategy for success may be to employ multi-functional therapeutics that target multiple PD-relevant pathogenic mechanisms. Evidence is presented that the multi-functional glycosphingolipid GM1 ganglioside may be just such a therapeutic.
Collapse
Affiliation(s)
- Jay S Schneider
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
14
|
Burtscher J, Pepe G, Maharjan N, Riguet N, Di Pardo A, Maglione V, Millet GP. Sphingolipids and impaired hypoxic stress responses in Huntington disease. Prog Lipid Res 2023; 90:101224. [PMID: 36898481 DOI: 10.1016/j.plipres.2023.101224] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/20/2023] [Accepted: 03/05/2023] [Indexed: 03/11/2023]
Abstract
Huntington disease (HD) is a debilitating, currently incurable disease. Protein aggregation and metabolic deficits are pathological hallmarks but their link to neurodegeneration and symptoms remains debated. Here, we summarize alterations in the levels of different sphingolipids in an attempt to characterize sphingolipid patterns specific to HD, an additional molecular hallmark of the disease. Based on the crucial role of sphingolipids in maintaining cellular homeostasis, the dynamic regulation of sphingolipids upon insults and their involvement in cellular stress responses, we hypothesize that maladaptations or blunted adaptations, especially following cellular stress due to reduced oxygen supply (hypoxia) contribute to the development of pathology in HD. We review how sphingolipids shape cellular energy metabolism and control proteostasis and suggest how these functions may fail in HD and in combination with additional insults. Finally, we evaluate the potential of improving cellular resilience in HD by conditioning approaches (improving the efficiency of cellular stress responses) and the role of sphingolipids therein. Sphingolipid metabolism is crucial for cellular homeostasis and for adaptations following cellular stress, including hypoxia. Inadequate cellular management of hypoxic stress likely contributes to HD progression, and sphingolipids are potential mediators. Targeting sphingolipids and the hypoxic stress response are novel treatment strategies for HD.
Collapse
Affiliation(s)
- Johannes Burtscher
- Institute of Sport Sciences, University of Lausanne, 1015 Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, 1005 Lausanne, Switzerland.
| | - Giuseppe Pepe
- IRCCS Neuromed, Via Dell'Elettronica, 86077 Pozzilli, Italy
| | - Niran Maharjan
- Department of Neurology, Center for Experimental Neurology, Inselspital University Hospital, 3010 Bern, Switzerland; Department for Biomedical Research (DBMR), University of Bern, 3010 Bern, Switzerland
| | | | - Alba Di Pardo
- IRCCS Neuromed, Via Dell'Elettronica, 86077 Pozzilli, Italy
| | | | - Grégoire P Millet
- Institute of Sport Sciences, University of Lausanne, 1015 Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, 1005 Lausanne, Switzerland
| |
Collapse
|
15
|
Chaudhari LR, Kawale AA, Desai SS, Kashte SB, Joshi MG. Pathophysiology of Spinal Cord Injury and Tissue Engineering Approach for Its Neuronal Regeneration: Current Status and Future Prospects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1409:51-81. [PMID: 36038807 DOI: 10.1007/5584_2022_731] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
A spinal cord injury (SCI) is a very debilitating condition causing loss of sensory and motor function as well as multiple organ failures. Current therapeutic options like surgery and pharmacotherapy show positive results but are incapable of providing a complete cure for chronic SCI symptoms. Tissue engineering, including neuroprotective or growth factors, stem cells, and biomaterial scaffolds, grabs attention because of their potential for regeneration and ability to bridge the gap in the injured spinal cord (SC). Preclinical studies with tissue engineering showed functional recovery and neurorestorative effects. Few clinical trials show the safety and efficacy of the tissue engineering approach. However, more studies should be carried out for potential treatment modalities. In this review, we summarize the pathophysiology of SCI and its current treatment modalities, including surgical, pharmacological, and tissue engineering approaches following SCI in preclinical and clinical phases.
Collapse
Affiliation(s)
- Leena R Chaudhari
- Department of Stem Cells and Regenerative Medicine, D. Y. Patil Education Society (Deemed to be University), Kolhapur, Maharashtra, India
| | - Akshay A Kawale
- Department of Stem Cells and Regenerative Medicine, D. Y. Patil Education Society (Deemed to be University), Kolhapur, Maharashtra, India
| | - Sangeeta S Desai
- Department of Obstetrics and Gynecology, Dr. D Y Patil Medical College, Hospital and Research Institute, Kolhapur, Maharashtra, India
| | - Shivaji B Kashte
- Department of Stem Cells and Regenerative Medicine, D. Y. Patil Education Society (Deemed to be University), Kolhapur, Maharashtra, India
| | - Meghnad G Joshi
- Department of Stem Cells and Regenerative Medicine, D. Y. Patil Education Society (Deemed to be University), Kolhapur, Maharashtra, India.
- Stem Plus Biotech, SMK Commercial Complex, Sangli, Maharashtra, India.
| |
Collapse
|
16
|
Vasques J, de Jesus Gonçalves R, da Silva-Junior A, Martins R, Gubert F, Mendez-Otero R. Gangliosides in nervous system development, regeneration, and pathologies. Neural Regen Res 2023. [PMID: 35799513 PMCID: PMC9241395 DOI: 10.4103/1673-5374.343890] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
17
|
Sandhoff R, Sandhoff K. Neuronal Ganglioside and Glycosphingolipid (GSL) Metabolism and Disease : Cascades of Secondary Metabolic Errors Can Generate Complex Pathologies (in LSDs). ADVANCES IN NEUROBIOLOGY 2023; 29:333-390. [PMID: 36255681 DOI: 10.1007/978-3-031-12390-0_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Glycosphingolipids (GSLs) are a diverse group of membrane components occurring mainly on the surfaces of mammalian cells. They and their metabolites have a role in intercellular communication, serving as versatile biochemical signals (Kaltner et al, Biochem J 476(18):2623-2655, 2019) and in many cellular pathways. Anionic GSLs, the sialic acid containing gangliosides (GGs), are essential constituents of neuronal cell surfaces, whereas anionic sulfatides are key components of myelin and myelin forming oligodendrocytes. The stepwise biosynthetic pathways of GSLs occur at and lead along the membranes of organellar surfaces of the secretory pathway. After formation of the hydrophobic ceramide membrane anchor of GSLs at the ER, membrane-spanning glycosyltransferases (GTs) of the Golgi and Trans-Golgi network generate cell type-specific GSL patterns for cellular surfaces. GSLs of the cellular plasma membrane can reach intra-lysosomal, i.e. luminal, vesicles (ILVs) by endocytic pathways for degradation. Soluble glycoproteins, the glycosidases, lipid binding and transfer proteins and acid ceramidase are needed for the lysosomal catabolism of GSLs at ILV-membrane surfaces. Inherited mutations triggering a functional loss of glycosylated lysosomal hydrolases and lipid binding proteins involved in GSL degradation cause a primary lysosomal accumulation of their non-degradable GSL substrates in lysosomal storage diseases (LSDs). Lipid binding proteins, the SAPs, and the various lipids of the ILV-membranes regulate GSL catabolism, but also primary storage compounds such as sphingomyelin (SM), cholesterol (Chol.), or chondroitin sulfate can effectively inhibit catabolic lysosomal pathways of GSLs. This causes cascades of metabolic errors, accumulating secondary lysosomal GSL- and GG- storage that can trigger a complex pathology (Breiden and Sandhoff, Int J Mol Sci 21(7):2566, 2020).
Collapse
Affiliation(s)
- Roger Sandhoff
- Lipid Pathobiochemistry Group, German Cancer Research Center, Heidelberg, Germany
| | - Konrad Sandhoff
- LIMES, c/o Kekule-Institute for Organic Chemistry and Biochemistry, University of Bonn, Bonn, Germany.
| |
Collapse
|
18
|
The Neuroprotective Effect of GM-1 Ganglioside on the Amyloid-Beta-Induced Oxidative Stress in PC-12 Cells Mediated by Nrf-2/ARE Signaling Pathway. Neurochem Res 2022; 47:2405-2415. [PMID: 35635605 DOI: 10.1007/s11064-022-03635-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is characterized by the accumulation of amyloid-β (Aβ) plaques, tau tangles, neuroinflammation, oxidative stress, and progressive memory deficits. Aβ deposition could exacerbate oxidative damage and cellular apoptosis. GM-1 ganglioside (GM-1) has previously been reported to exhibit neuroprotective effects in rodents and patients with AD. However, the substantial impacts and mechanism of GM-1 on Aβ-induced oxidative stress remain elusive. The present study used PC-12 pheochromocytoma cells treated with Aβ25-35 peptide to construct the AD model in vitro. Aβ25-35 administration alone inhibited cell viability and facilitated cell apoptosis in the range doses of 10 μM to 30 μM. At the same time, GM-1 supplementation promoted cell proliferation and rescued cell apoptosis in a dose-dependent fashion ranging from 5 to 30 μM. In parallel, GM-1 treatment alleviated Aβ-induced oxidative stress by increasing the level of antioxidant enzymes and decreasing the content of malondialdehyde (MDA). The nuclear factor-E2-related factor 2 (Nrf2) is a crucial mediator of antioxidant response. We reported herein that GM-1 could activate Nrf-2 in the PC-12 cells co-treated with Aβ25-35, following with the activated expression of antioxidant response elements (ARE)-mediated antioxidant and detoxifying genes. Consistently, knock-down of Nrf-2 via siRNA abolished the beneficial decrease of Aβ-induced oxidative stress by GM-1 treatment, indicating that GM-1-improved oxidative stress was regulated by the Nrf-2 signaling pathway. Collectively, GM-1 could alleviate Aβ25-35-induced oxidative damage mediated through the Nrf-2/ARE signaling pathway, which might be a potential agent for AD treatment.
Collapse
|
19
|
Wang Q, Duan L, Li X, Wang Y, Guo W, Guan F, Ma S. Glucose Metabolism, Neural Cell Senescence and Alzheimer’s Disease. Int J Mol Sci 2022; 23:ijms23084351. [PMID: 35457168 PMCID: PMC9030802 DOI: 10.3390/ijms23084351] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/05/2022] [Accepted: 04/12/2022] [Indexed: 12/20/2022] Open
Abstract
Alzheimer’s disease (AD), an elderly neurodegenerative disorder with a high incidence and progressive memory decline, is one of the most expensive, lethal, and burdening diseases. To date, the pathogenesis of AD has not been fully illustrated. Emerging studies have revealed that cellular senescence and abnormal glucose metabolism in the brain are the early hallmarks of AD. Moreover, cellular senescence and glucose metabolism disturbance in the brain of AD patients may precede amyloid-β deposition or Tau protein phosphorylation. Thus, metabolic reprogramming targeting senescent microglia and astrocytes may be a novel strategy for AD intervention and treatment. Here, we recapitulate the relationships between neural cell senescence and abnormal glucose metabolism (e.g., insulin signaling, glucose and lactate metabolism) in AD. We then discuss the potential perspective of metabolic reprogramming towards an AD intervention, providing a theoretical basis for the further exploration of the pathogenesis of and therapeutic approach toward AD.
Collapse
Affiliation(s)
- Qianqian Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (Q.W.); (L.D.); (X.L.); (Y.W.); (W.G.)
| | - Linyan Duan
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (Q.W.); (L.D.); (X.L.); (Y.W.); (W.G.)
| | - Xingfan Li
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (Q.W.); (L.D.); (X.L.); (Y.W.); (W.G.)
| | - Yifu Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (Q.W.); (L.D.); (X.L.); (Y.W.); (W.G.)
| | - Wenna Guo
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (Q.W.); (L.D.); (X.L.); (Y.W.); (W.G.)
| | - Fangxia Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (Q.W.); (L.D.); (X.L.); (Y.W.); (W.G.)
- Institute of Neuroscience, Zhengzhou University, Zhengzhou 450052, China
- NHC Key Laboratory of Birth Defects Prevention, Henan Institute of Reproduction Health Science and Technology, Zhengzhou 450002, China
- Correspondence: (F.G.); (S.M.)
| | - Shanshan Ma
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (Q.W.); (L.D.); (X.L.); (Y.W.); (W.G.)
- Institute of Neuroscience, Zhengzhou University, Zhengzhou 450052, China
- NHC Key Laboratory of Birth Defects Prevention, Henan Institute of Reproduction Health Science and Technology, Zhengzhou 450002, China
- Correspondence: (F.G.); (S.M.)
| |
Collapse
|
20
|
Recent behavioral findings of pathophysiological involvement of lactate in the central nervous system. Biochim Biophys Acta Gen Subj 2022; 1866:130137. [DOI: 10.1016/j.bbagen.2022.130137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 11/19/2022]
|
21
|
Yu G, Zhang Y, Ning B. Reactive Astrocytes in Central Nervous System Injury: Subgroup and Potential Therapy. Front Cell Neurosci 2022; 15:792764. [PMID: 35002629 PMCID: PMC8733560 DOI: 10.3389/fncel.2021.792764] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022] Open
Abstract
Traumatic central nervous system (CNS) injury, which includes both traumatic brain injury (TBI) and spinal cord injury (SCI), is associated with irreversible loss of neurological function and high medical care costs. Currently, no effective treatment exists to improve the prognosis of patients. Astrocytes comprise the largest population of glial cells in the CNS and, with the advancements in the field of neurology, are increasingly recognized as having key functions in both the brain and the spinal cord. When stimulated by disease or injury, astrocytes become activated and undergo a series of changes, including alterations in gene expression, hypertrophy, the loss of inherent functions, and the acquisition of new ones. Studies have shown that astrocytes are highly heterogeneous with respect to their gene expression profiles, and this heterogeneity accounts for their observed context-dependent phenotypic diversity. In the inured CNS, activated astrocytes play a dual role both as regulators of neuroinflammation and in scar formation. Identifying the subpopulations of reactive astrocytes that exert beneficial or harmful effects will aid in deciphering the pathological mechanisms underlying CNS injuries and ultimately provide a theoretical basis for the development of effective strategies for the treatment of associated conditions. Following CNS injury, as the disease progresses, astrocyte phenotypes undergo continuous changes. Although current research methods do not allow a comprehensive and accurate classification of astrocyte subpopulations in complex pathological contexts, they can nonetheless aid in understanding the roles of astrocytes in disease. In this review, after a brief introduction to the pathology of CNS injury, we summarize current knowledge regarding astrocyte activation following CNS injury, including: (a) the regulatory factors involved in this process; (b) the functions of different astrocyte subgroups based on the existing classification of astrocytes; and (c) attempts at astrocyte-targeted therapy.
Collapse
Affiliation(s)
- GuiLian Yu
- Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ying Zhang
- Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bin Ning
- Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
22
|
Role of Receptors in Relation to Plaques and Tangles in Alzheimer's Disease Pathology. Int J Mol Sci 2021; 22:ijms222312987. [PMID: 34884789 PMCID: PMC8657621 DOI: 10.3390/ijms222312987] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/26/2021] [Accepted: 11/28/2021] [Indexed: 12/23/2022] Open
Abstract
Despite the identification of Aβ plaques and NFTs as biomarkers for Alzheimer’s disease (AD) pathology, therapeutic interventions remain elusive, with neither an absolute prophylactic nor a curative medication available to impede the progression of AD presently available. Current approaches focus on symptomatic treatments to maintain AD patients’ mental stability and behavioral symptoms by decreasing neuronal degeneration; however, the complexity of AD pathology requires a wide range of therapeutic approaches for both preventive and curative treatments. In this regard, this review summarizes the role of receptors as a potential target for treating AD and focuses on the path of major receptors which are responsible for AD progression. This review gives an overall idea centering on major receptors, their agonist and antagonist and future prospects of viral mimicry in AD pathology. This article aims to provide researchers and developers a comprehensive idea about the different receptors involved in AD pathogenesis that may lead to finding a new therapeutic strategy to treat AD.
Collapse
|