1
|
Bonfili L, Grasselli FM, Cuccioloni M, Cecarini V, Lufrano D, Vittadini E, Galosi L, Sonsini G, Ubaldi M, Turck JL, Medina LFDC, Suchodolski J, Eleuteri AM. A red lentils-based synbiotic cookie exerts neuroprotective effects in a mouse model of Alzheimer's disease. J Nutr Biochem 2025; 141:109904. [PMID: 40120777 DOI: 10.1016/j.jnutbio.2025.109904] [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: 01/24/2025] [Revised: 03/04/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
Gut microbiota preservation or rational manipulation is a key condition for healthy longevity and a promising strategy to prevent neurodegenerations exploiting the gut-brain axis, with a key role of prebiotics and probiotics. Whether their combination in a functional food can provide a synergistic effect to the host remains controversial. To fill this gap, we supplemented the diet of 3xTg-AD Alzheimer's disease mice with a red lentils (prebiotic)-based cookie enriched with neuroprotective probiotics and we performed behavioural, biochemical and molecular tests. Chronic consumption of this synbiotic preparation (functional cookie) preserved cognition, reduced amyloid load, improved glucose and lipid homeostasis and diminished oxidation and inflammation related damages compared to animals receiving a classic cookie (standard recipe). The synergistic effect was indicated by significantly higher glucose insulinotropic polypeptide concentrations in the functional cookie group compared to probiotic group. Moreover, Ruminoclostridium sp KB18 and Ruminicoccus decreased in the gut of mice supplemented with the functional cookie, partially explaining the improved short-term memory upon treatments and substantiating the combined use over individual components. This synbiotic innovative snack represents a prototype of a simple and affordable dietary approach to promote healthy aging and prevent or delay the onset of neurodegenerations.
Collapse
Affiliation(s)
- Laura Bonfili
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, MC, Italy.
| | | | | | - Valentina Cecarini
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, MC, Italy
| | - Daniela Lufrano
- Department of Biological sciences, Faculty of Exact Sciences, National University of La Plata (UNLP) B1900 La Plata, Buenos Aires, Argentina
| | - Elena Vittadini
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, MC, Italy
| | - Livio Galosi
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, MC, Italy
| | | | - Massimo Ubaldi
- School of Pharmacy, University of Camerino, Camerino, MC, Italy
| | | | | | - Jan Suchodolski
- Gastrointestinal Laboratory, Texas A&M University, College Station, TX, USA
| | - Anna Maria Eleuteri
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, MC, Italy.
| |
Collapse
|
2
|
Chen L, Jiang H, Licinio J, Wu H. Brain O-GlcNAcylation: Bridging physiological functions, disease mechanisms, and therapeutic applications. Mol Psychiatry 2025; 30:2754-2772. [PMID: 40033044 PMCID: PMC12092303 DOI: 10.1038/s41380-025-02943-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/13/2025] [Accepted: 02/25/2025] [Indexed: 03/05/2025]
Abstract
O-GlcNAcylation, a dynamic post-translational modification occurring on serine or threonine residues of numerous proteins, plays a pivotal role in various cellular processes, including gene regulation, metabolism, and stress response. Abundant in the brain, O-GlcNAcylation intricately governs neurodevelopment, synaptic assembly, and neuronal functions. Recent investigations have established a correlation between the dysregulation of brain O-GlcNAcylation and a broad spectrum of neurological disorders and injuries, spanning neurodevelopmental, neurodegenerative, and psychiatric conditions, as well as injuries to the central nervous system (CNS). Manipulating O-GlcNAcylation has demonstrated neuroprotective properties against these afflictions. This review delineates the roles and mechanisms of O-GlcNAcylation in the CNS under both physiological and pathological circumstances, with a focus on its neuroprotective effects in neurological disorders and injuries. We discuss the involvement of O-GlcNAcylation in key processes such as neurogenesis, synaptic plasticity, and energy metabolism, as well as its implications in conditions like Alzheimer's disease, Parkinson's disease, and ischemic stroke. Additionally, we explore prospective therapeutic approaches for CNS disorders and injuries by targeting O-GlcNAcylation, highlighting recent clinical developments and future research directions. This comprehensive overview aims to provide insights into the potential of O-GlcNAcylation as a therapeutic target and guide future investigations in this promising field.
Collapse
Affiliation(s)
- Liping Chen
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Huihui Jiang
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Julio Licinio
- Department of Psychiatry, Norton College of Medicine, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA
| | - Haitao Wu
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China.
- Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, China.
- Chinese Institute for Brain Research, Beijing, 102206, China.
| |
Collapse
|
3
|
Zhao S, Zheng Y. Neuroplasticity After Hypoxic-Ischemic Brain Injury in Neonatal Pigs Based on Time-Dependent Behavior of 1H-MRS-Tau Protein and Synaptic Associated Proteins and Synaptic Structure Analysis. Neurochem Res 2025; 50:169. [PMID: 40407938 PMCID: PMC12102000 DOI: 10.1007/s11064-025-04421-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 05/09/2025] [Accepted: 05/12/2025] [Indexed: 05/26/2025]
Abstract
This study investigated the effects of hypoxic-ischemic (HI) injury on neonatal neuroplasticity using the following approaches: Magnetic Resonance Spectroscopy (1H-MRS) imaging to analyze dynamic changes in tau protein levels, immunofluorescence staining to evaluate synaptophysin (SYP), neurocan (Neu), and tau protein, and utilizing transmission electron microscopy (TEM) to examine synaptic ultrastructure at multiple time points. A total of 59 healthy neonatal pigs were included, with 10 in the control group and 43 in the HI model group. The results demonstrated that SYP immunostaining intensity peaked at 6-12 h after HI before declining. Neu expression exhibited an initial decrease, followed by a transient increase and subsequent reduction, reaching its lowest level at 6-12 h after HI. Tau protein levels increased initially after HI, peaked at 24-48 h after HI, and subsequently decreased. SYP was negatively correlated with Neu with a correlation coefficient of -0.877. SYP was not correlated with Tau, neither was Neu with Tau. Compared with the control group, the number of synaptic vesicles decreased, and Post-Synaptic Density (PSD) thickness increased 6-12 h after HI. At 12-24 h after HI, the number of synaptic vesicles increased, and PSD thickness slightly decreased. At 24-48 h after HI, the vesicle number decreased, PSD became thinner, interrupting continuity, mitochondria swelled, and mitochondrial cristae blurred and disappeared. The findings suggest that the expression of Tau, SYP, and Neu is linked to alterations in synaptic and myelin structures, reflecting varying aspects of neural plasticity following HI injury.
Collapse
Affiliation(s)
- Sijia Zhao
- Department of Radiology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, PR China
| | - Yang Zheng
- Department of Radiology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, PR China.
| |
Collapse
|
4
|
Azmoun S, Lewis F, Shoieb D, Jin Y, Colicino E, Winters I, Gu H, Krishnamurthy H, Richardson J, Placidi D, Lambertini L, Lucchini RG. Impact of Manganese on Neuronal Function: An Exploratory MultiOmic Study on Ferroalloy Workers in Brescia, Italy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.05.02.25326824. [PMID: 40385409 PMCID: PMC12083615 DOI: 10.1101/2025.05.02.25326824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Interest is growing in the potential role of manganese (Mn) in Alzheimer's Disease (ADRD). This nested pilot study of a ferroalloy workers cohort was aimed to investigate the effects of long-term occupational Mn exposure on cognitive function through β-amyloid (Aβ) modification and brain deposition, as well as metabolomic, lipidomic and proteomic profiling. We examined 6 male exposed workers (median age 63, exposure duration 31 yrs), and 5 historical controls (median age 60) who had undergone brain PET scan imaging showing higher Aβ deposition among the exposed compared to the controls (p < 0.05). The average annual cumulative respirable Mn of the ferroalloy workers was 329.23 ± 516.39 μg/m3 (geometric mean 118.59). Average Mn level in plasma of the exposed subjects (0.704 ± 0.2 ng/mL) was significantly higher than the controls (0.397 ± 0.18). Pathway analyses using LC-MS/MS results revealed impacted metabolomic pathways such as olfactory signaling, mitochondrial fatty acid beta-oxidation, biogenic amine synthesis, SLC-mediated transmembrane transport, and glycerophospholipid and choline metabolism in the Mn exposed group. Single molecule arrays (Simoa) analysis revealed notable modifications of AD-related plasma biomarkers; protein microarray (chip) showed significant changes (p < 0.05) in the levels of some plasma antibodies targeting autoimmune and neuronal associated proteins such as Aβ (25-35), GFAP, Serotonin, Human NOVA1, and Human Siglec-1/CD169 among the Mn exposed individuals. This data provides evidence on Mn-induced alterations of pathways and biomarkers associated with cognitive neurodegenerative diseases.
Collapse
Affiliation(s)
- Somaiyeh Azmoun
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social work, Florida International University, Miami, FL, USA
| | - Freeman Lewis
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social work, Florida International University, Miami, FL, USA
| | - Daniel Shoieb
- Department of Medical and Surgical Specialties, University of Brescia, Brescia, Italy
| | - Yan Jin
- St. Jude Children Research Hospital, Memphis, TN, USA
| | - Elena Colicino
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Isha Winters
- Isakson Center for Neurological Disease Research and Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, GA, USA
| | - Haiwei Gu
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | | | - Jason Richardson
- Isakson Center for Neurological Disease Research and Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, GA, USA
| | - Donatella Placidi
- Department of Medical and Surgical Specialties, University of Brescia, Brescia, Italy
| | - Luca Lambertini
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Roberto G Lucchini
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social work, Florida International University, Miami, FL, USA
- Occupational Medicine, University of Modena and Reggio Emilia, Italy
| |
Collapse
|
5
|
Mirabelli M, Chiefari E, Arcidiacono B, Salatino A, Pascarella A, Morelli M, Credendino SC, Brunetti FS, Di Vito A, Greco A, Huin V, Nicoletti F, Pierantoni GM, Fedele M, Aguglia U, Foti DP, Brunetti A. HMGA1 deficiency: a pathogenic link between tau pathology and insulin resistance. EBioMedicine 2025; 115:105700. [PMID: 40233659 PMCID: PMC12019291 DOI: 10.1016/j.ebiom.2025.105700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/29/2025] [Accepted: 04/01/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Growing evidence links tau-related neurodegeneration with insulin resistance and type 2 diabetes (T2D), though the underlying mechanisms remain unclear. Our previous research identified HMGA1 as crucial for insulin receptor (INSR) expression, with defects in the HMGA1 gene associated with insulin resistance and T2D. Here, we explore HMGA1 deficiency as a potential contributor to tauopathies, such as Alzheimer's disease (AD), and its connection to insulin resistance. METHODS Immunoblot analyses, protein-DNA interaction studies, ChIP-qPCR, and reporter gene assays were conducted in human and mouse neuronal cell models. Tau immunohistochemistry, behavioural studies, and brain glucose metabolism were analysed in Hmga1-knockout mice. Additionally, a case-control study investigated the relationship between HMGA1 and tau pathology in patients with tauopathy, carrying or not the HMGA1 rs146052672 variant, known to reduce HMGA1 protein levels and increase the risk of insulin resistance and T2D. FINDINGS We show that HMGA1 regulates tau protein expression primarily through the specific repression of MAPT gene transcription. In both human neuronal cells and primary mouse neurons, tau mRNA and protein levels were inversely correlated with HMGA1 expression. This inverse relationship was further confirmed in the brain of Hmga1-knockout mice, where tau was overexpressed, INSR was downregulated, and brain glucose uptake was impaired. Additionally, the rs146052672 variant was more common in patients with tauopathy (12/69, 17.4%) than in controls (10/200, 5.0%) (p = 0.001), and carriers of this variant exhibited more severe disease progression and poorer therapeutic outcomes. INTERPRETATION These findings suggest that HMGA1 deficiency may drive tau pathology, linking tauopathies to insulin resistance and providing new insights into the relationship between metabolic and neurodegenerative disorders. Furthermore, our observation that over 17% of individuals with tauopathy exhibit a deficit in HMGA1 protein production could have significant clinical implications, potentially guiding the development of therapeutic strategies targeting this specific defect. FUNDING See acknowledgements section.
Collapse
Affiliation(s)
- Maria Mirabelli
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Eusebio Chiefari
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Biagio Arcidiacono
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Alessandro Salatino
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Angelo Pascarella
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Maurizio Morelli
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Sara C Credendino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Francesco S Brunetti
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Anna Di Vito
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Adelaide Greco
- Interdepartmental Centre of Veterinary Radiology, University of Naples "Federico II", Naples, Italy
| | - Vincent Huin
- University of Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, UMR-S1172, Team Alzheimer & Tauopathies, F-59000, Lille, France
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, University of Rome "Sapienza", Rome, Italy; IRCCS Neuromed, Pozzilli, Italy
| | - Giovanna M Pierantoni
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Monica Fedele
- Institute of Experimental Endocrinology and Oncology, CNR, Naples, Italy
| | - Umberto Aguglia
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Daniela P Foti
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Græcia", Catanzaro, Italy.
| | - Antonio Brunetti
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy.
| |
Collapse
|
6
|
Sharma V, Verma R, Singh TG. Targeting hypoxia-related pathobiology in Alzheimer's disease: strategies for prevention and treatment. Mol Biol Rep 2025; 52:416. [PMID: 40266407 DOI: 10.1007/s11033-025-10520-4] [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: 07/16/2024] [Accepted: 04/15/2025] [Indexed: 04/24/2025]
Abstract
INTRODUCTION Alzheimer's Disease (AD) is a neurodegenerative condition characterised by cognitive decline and memory impairment. Recent research highlights the important role of hypoxia, a state of insufficient oxygen availability, in exacerbating AD pathogenesis. MATERIALS AND METHODS Through the use of a number of different search engines like Scopus, PubMed, Bentham, and Elsevier databases, a literature review was carried out for investigating the role of hypoxia mediated pathobiology in AD. Only peerreviewed articles published in reputable journals in English language were included. Conversely, non-peer-reviewed articles, conference abstracts, and editorials were excluded, along with studies lacking experimental or clinical relevance or those unavailable in full text. CONCLUSION Hypoxia exacerbates core pathological features such as oxidative stress, neuroinflammation, mitochondrial dysfunction, amyloid-beta (Aβ) dysregulation, and hyperphosphorylation of tau protein. These interlinked mechanisms establish a self-perpetuating cycle of neuronal damage, accelerating disease progression. Addressing hypoxia as a modifiable risk factor offers potential for both prevention and treatment of AD. Exploring hypoxia and the HIF signalling pathway may help counteract the neuropathological and symptomatic effects of neurodegeneration.
Collapse
Affiliation(s)
- Veerta Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Reet Verma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| |
Collapse
|
7
|
Onkar A, Sheshadri D, Nagarajan K, Ganesh S. Inactivation of Laforin Phosphatase and Increased Glucose Uptake Underlie Glycogen Synthase-Mediated Neuronal Survival Under Oxidative Stress. Mol Neurobiol 2025:10.1007/s12035-025-04955-w. [PMID: 40261604 DOI: 10.1007/s12035-025-04955-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 04/13/2025] [Indexed: 04/24/2025]
Abstract
Recent studies demonstrate that exposure of neurons to physiological stressors triggers glycogen synthase (GS) activation and glycogen synthesis as a transient cell survival mechanism. However, the mechanisms that regulate glycogen synthesis during stress and its role in neuronal physiology remain unclear. This study investigated the mechanisms that guide GS activation and glycogen accumulation under oxidative stress conditions as a model stressor. We use neuronal cell lines to demonstrate that hydrogen peroxide-induced oxidative stress activates GS and glycogen synthesis in neuronal cells. We further demonstrate that the stress-induced glycogen accumulation is dependent on the membrane localization of the Glut3 glucose transporters and increased glucose uptake during stress. The stress-induced activation of glycogen synthesis, however, is independent of intracellular glucose level, suggesting a parallel mechanism for activating GS and glucose uptake in neurons under physiological stress. We demonstrate that oxidative stress results in the inactivation of laforin phosphatase, leading to the membrane localization of Glut3 and activation of GS. Using the Drosophila model, we demonstrate that increased GS activity and concomitant glycogen accumulation are pro-survival mechanisms for neurons under oxidative stress. Our study thus offers novel insights into the pathways that regulate glycogen metabolism in neurons under oxidative stress and underscores their importance for neuronal survival.
Collapse
Affiliation(s)
- Akanksha Onkar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, India
- Current address: Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Deepashree Sheshadri
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, India
- Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kanpur, 208016, India
| | - Kamali Nagarajan
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, India
- Current address: Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Subramaniam Ganesh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, India.
- Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, 208016, India.
| |
Collapse
|
8
|
Li N, Fang X, Li H, Liu J, Chen N, Zhao X, Yang Q, Chen X. Ginsenoside CK modulates glucose metabolism via PPARγ to ameliorate SCOP-induced cognitive dysfunction. Metab Brain Dis 2025; 40:168. [PMID: 40178645 DOI: 10.1007/s11011-025-01596-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 03/23/2025] [Indexed: 04/05/2025]
Abstract
Ginsenoside compound K (CK) exhibits neuroprotective properties; however, the underlying mechanisms behind these effects have not been investigated thoroughly. CK is the primary active compound derived from ginseng and is metabolized in the gut. It enhances neuronal function by modulating the gut microflora. Therefore, the present study aimed to elucidate the mechanism through which CK enhances cognitive function, employing gut microbiome and microarray analyses. The results revealed that CK upregulated the expression of peroxisome proliferator-activated receptor gamma (PPARγ), suppressed amyloid-β (Aβ) aggregation in hippocampal neurons, and influenced the expression of cyclin-dependent kinase-5 (CDK5), (including insulin receptor substrate 2) IRS2, insulin-degrading enzyme (IDE), glycogen synthase kinase-3 beta (GSK-3β), glucose transporter type 1 (GLUT1), and glucose transporter type 3 (GLUT3) proteins. These proteins play crucial roles in regulating brain glucose metabolism, increasing neuronal energy, and reducing neuronal apoptosis, thereby ameliorating cognitive impairment in mice.
Collapse
Affiliation(s)
- Na Li
- Jinlin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, P.R. China
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore
| | - Xingyu Fang
- Jinlin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, P.R. China
| | - Hui Li
- Qian Wei Hospital of Jilin Province, Changchun, 130117, Jilin, P.R. China
| | - Jian Liu
- Jinlin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, P.R. China
| | - Nan Chen
- Jinlin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, P.R. China
| | - Xiaohui Zhao
- Jinlin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, P.R. China
| | - Qing Yang
- Jinlin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, P.R. China.
| | - Xijun Chen
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, P.R. China.
| |
Collapse
|
9
|
Oka M, Nakajima S, Suzuki E, Yamamoto S, Ando K. Glucose uptake in pigment glia suppresses Tau-induced inflammation and photoreceptor degeneration. Dis Model Mech 2025; 18:dmm052057. [PMID: 40151148 PMCID: PMC12067088 DOI: 10.1242/dmm.052057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
Brain inflammation contributes to the pathogenesis of neurodegenerative diseases, such as Alzheimer's disease (AD). Glucose hypometabolism and glial activation are pathological features seen in AD brains; however, the connection between the two is not fully understood. Using a Drosophila model of AD, we identified that glucose metabolism in glia plays a critical role in neuroinflammation under disease conditions. Expression of human MATP (hereafter referred to as Tau) in the retinal cells, including photoreceptor neurons and pigment glia, causes photoreceptor degeneration accompanied by the formation of dark-stained round inclusion-like structures and swelling of the lamina cortex. We found that inclusion-like structures are formed by glial phagocytosis, and swelling of the laminal cortex correlates with the expression of antimicrobial peptides. Coexpression of human glucose transporter 3 (SLC2A3, hereafter referred to as GLUT3) with Tau in the retina does not affect Tau levels but suppresses these inflammatory responses and photoreceptor degeneration. We also found that expression of GLUT3, specifically in the pigment glia, is sufficient to suppress inflammatory phenotypes and mitigate photoreceptor degeneration in the Tau-expressing retina. Our results suggest that glial glucose metabolism contributes to inflammatory responses and neurodegeneration in tauopathy.
Collapse
Affiliation(s)
- Mikiko Oka
- Department of Biological Sciences, Tokyo Metropolitan University, Tokyo 192-0397, Japan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute for Texas Children's Hospital, Houston, TX 77030, USA
| | - Sho Nakajima
- Department of Biological Sciences, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Emiko Suzuki
- Department of Biological Sciences, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute for Texas Children's Hospital, Houston, TX 77030, USA
| | - Kanae Ando
- Department of Biological Sciences, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| |
Collapse
|
10
|
Guzmán‐Hernández R, Fossati S. Fibrillar tau alters cerebral endothelial cell metabolism, vascular inflammatory activation, and barrier function in vitro and in vivo. Alzheimers Dement 2025; 21:e70077. [PMID: 40110691 PMCID: PMC11923556 DOI: 10.1002/alz.70077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/10/2025] [Accepted: 02/10/2025] [Indexed: 03/22/2025]
Abstract
INTRODUCTION The presence of tau aggregates in and around the brain vasculature in Alzheimer's disease (AD) and tauopathies suggests its possible pathogenicity to cerebral endothelial cells (ECs). METHODS We used an in vitro model of the blood-brain barrier (BBB) to understand the mechanisms of fibrillar tau-mediated cerebral EC and BBB pathology, confirming our findings in 3-month-old P301S mice brains and extracted microvessels. RESULTS Protofibrillar and fibrillar tau species induce endothelial barrier permeability through an increase in glycolysis, which activates ECs toward a pro-inflammatory phenotype, inducing loss of junction protein expression and localization. The Warburg-like metabolic shift toward glycolysis and increased vascular pathological phenotypes are also present in young P301S mice. DISCUSSION In sum, our work reveals that fibrillar tau species, by enhancing endothelial glycolytic metabolism, promote vascular inflammatory phenotypes and loss of BBB function, highlighting the importance of addressing and targeting early tau-mediated neurovascular damage in AD and tauopathies. HIGHLIGHTS We improve the understanding of the mechanisms of vascular pathology in tauopathies. Fibrillar tau mediates vascular metabolic changes, inflammation, and blood-brain barrier (BBB) dysfunction. These events are replicated at early stages in a tauopathy mouse model. Inhibiting altered glycolysis reduces BBB permeability and endothelial activation.
Collapse
Affiliation(s)
- Roberto Guzmán‐Hernández
- Alzheimer's Center at TempleDepartment of Neural SciencesTemple University Lewis Katz School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Silvia Fossati
- Alzheimer's Center at TempleDepartment of Neural SciencesTemple University Lewis Katz School of MedicinePhiladelphiaPennsylvaniaUSA
| |
Collapse
|
11
|
Oka M, Nakajima S, Suzuki E, Yamamoto S, Ando K. Glucose uptake in pigment glia suppresses tau-induced inflammation and photoreceptor degeneration in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.14.607919. [PMID: 39229232 PMCID: PMC11370381 DOI: 10.1101/2024.08.14.607919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Brain inflammation contributes to the pathogenesis of neurodegenerative diseases such as Alzheimer's disease (AD). Glucose hypometabolism and glial activation are pathological features seen in AD brains; however, the connection between the two is not fully understood. Using a Drosophila model of AD, we identified that glucose metabolism in glia plays a critical role in neuroinflammation under disease conditions. Expression of human Tau in the retinal cells, including photoreceptor neurons and pigment glia, causes photoreceptor degeneration accompanied by inclusion formation and swelling of the lamina cortex. We found that inclusions are formed by glial phagocytosis, and swelling of the laminal cortex correlates with the expression of antimicrobial peptides. Co-expression of human glucose transporter 3 ( GLUT3 ) with Tau in the retina does not affect tau levels but suppresses these inflammatory responses and photoreceptor degeneration. We also found that expression of GLUT3 , specifically in the pigment glia, is sufficient to suppress inflammatory phenotypes and mitigate photoreceptor degeneration in the tau-expressing retina. Our results suggest that glial glucose metabolism contributes to inflammatory responses and neurodegeneration in tauopathy. Summary Statement Glucose uptake into pigment glia suppresses inflammatory responses and photoreceptor degeneration in the fly model of tauopathy.
Collapse
|
12
|
Rojas-Pirela M, Andrade-Alviárez D, Rojas V, Marcos M, Salete-Granado D, Chacón-Arnaude M, Pérez-Nieto MÁ, Kemmerling U, Concepción JL, Michels PAM, Quiñones W. Exploring glycolytic enzymes in disease: potential biomarkers and therapeutic targets in neurodegeneration, cancer and parasitic infections. Open Biol 2025; 15:240239. [PMID: 39904372 PMCID: PMC11793985 DOI: 10.1098/rsob.240239] [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: 08/26/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 02/06/2025] Open
Abstract
Glycolysis, present in most organisms, is evolutionarily one of the oldest metabolic pathways. It has great relevance at a physiological level because it is responsible for generating ATP in the cell through the conversion of glucose into pyruvate and reducing nicotinamide adenine dinucleotide (NADH) (that may be fed into the electron chain in the mitochondria to produce additional ATP by oxidative phosphorylation), as well as for producing intermediates that can serve as substrates for other metabolic processes. Glycolysis takes place through 10 consecutive chemical reactions, each of which is catalysed by a specific enzyme. Although energy transduction by glucose metabolism is the main function of this pathway, involvement in virulence, growth, pathogen-host interactions, immunomodulation and adaptation to environmental conditions are other functions attributed to this metabolic pathway. In humans, where glycolysis occurs mainly in the cytosol, the mislocalization of some glycolytic enzymes in various other subcellular locations, as well as alterations in their expression and regulation, has been associated with the development and progression of various diseases. In this review, we describe the role of glycolytic enzymes in the pathogenesis of diseases of clinical interest. In addition, the potential role of these enzymes as targets for drug development and their potential for use as diagnostic and prognostic markers of some pathologies are also discussed.
Collapse
Affiliation(s)
- Maura Rojas-Pirela
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca37007, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
- Servicio de Medicina Interna, Hospital Universitario de Salamanca, Salamanca37007, Spain
| | - Diego Andrade-Alviárez
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| | - Verónica Rojas
- Instituto de Biología, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso2373223, Chile
| | - Miguel Marcos
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca37007, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
- Servicio de Medicina Interna, Hospital Universitario de Salamanca, Salamanca37007, Spain
| | - Daniel Salete-Granado
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca37007, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
| | - Marirene Chacón-Arnaude
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| | - María Á. Pérez-Nieto
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
- Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León, Soria42002, Spain
| | - Ulrike Kemmerling
- Instituto de Ciencias Biomédicas, Universidad de Chile, Facultad de Medicina, Santiago de Chile8380453, Chile
| | - Juan Luis Concepción
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| | - Paul A. M. Michels
- School of Biological Sciences, University of Edinburgh, The King’s Buildings, EdinburghEH9 3FL, UK
| | - Wilfredo Quiñones
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| |
Collapse
|
13
|
Manna PR, Yang S, Manna C, Waters H, Islam MA, Reddy AP, Rawat P, Reddy PH. Steroidogenic acute regulatory protein mediated variations of gender-specific sex neurosteroids in Alzheimer's disease: Relevance to hormonal and neuronal imbalance. Neurosci Biobehav Rev 2025; 169:105969. [PMID: 39631487 DOI: 10.1016/j.neubiorev.2024.105969] [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: 05/13/2024] [Revised: 07/24/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
The steroidogenic acute regulatory (StAR) protein mediates the rate-liming step in neuro/steroid biosynthesis. Multifaceted and delicate changes during aging, disrupting hormonal and neuronal homeostasis, constitute human senescence, an inevitable phenomenon that attributes to increased morbidity and mortality. Aging, along with progressive decreases in bioactive neurosteroids, is the primary risk factor for Alzheimer's disease (AD), which preferentially impacts two-thirds of women and one-third of men. AD is neuropathologically characterized by the accumulation of extracellular amyloid-β and intracellular phosphorylated Tau containing neurofibrillary tangles, resulting in dementia. Postmortem brains pertaining to gender-specific AD patients exhibit varied suppression of StAR and sex neurosteroid levels compared with age-matched cognitively healthy subjects, in which the attenuation of StAR is inversely correlated with the AD pathological markers. Interestingly, retinoid signaling upregulates StAR-motivated neurosteroid biosynthesis and reinstates various neurodegenerative vulnerabilities that promote AD pathogenesis. This review summarizes current understanding of StAR-driven alterations of sex neurosteroids in gender-specific AD risks and provides biochemical and molecular insights into therapeutic interventions for preventing and/or alleviating dementia for healthy aging.
Collapse
Affiliation(s)
- Pulak R Manna
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Shengping Yang
- Department of Biostatistics, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA
| | - Chayan Manna
- Baylor College of Medicine, Ben Taub Research Center, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Hope Waters
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Md Ariful Islam
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Arubala P Reddy
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Priyanka Rawat
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA; Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
14
|
Beltrán FA, Torres-Díaz L L, Troncoso-Escudero P, Villalobos-González J, Mayorga-Weber G, Lara M, Covarrubias-Pinto A, Valdivia S, Vicencio I, Papic E, Paredes-Martínez C, Silva-Januàrio ME, Rojas A, daSilva LLP, Court F, Rosas-Arellano A, Bátiz LF, Rojas P, Rivera FJ, Castro MA. Distinct roles of ascorbic acid in extracellular vesicles and free form: Implications for metabolism and oxidative stress in presymptomatic Huntington's disease. Free Radic Biol Med 2025; 227:521-535. [PMID: 39662690 DOI: 10.1016/j.freeradbiomed.2024.12.001] [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: 10/23/2024] [Revised: 11/18/2024] [Accepted: 12/02/2024] [Indexed: 12/13/2024]
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by a CAG trinucleotide repeat expansion in the first exon of the huntingtin gene. The huntingtin protein (Htt) is ubiquitously expressed and localized in several organelles, including endosomes, where it plays an essential role in intracellular trafficking. Presymptomatic HD is associated with a failure in energy metabolism and oxidative stress. Ascorbic acid is a potent antioxidant that plays a key role in modulating neuronal metabolism and is highly concentrated in the brain. During synaptic activity, neurons take up ascorbic acid released by glial cells; however, this process is disrupted in HD. In this study, we aim to elucidate the molecular and cellular mechanisms underlying this dysfunction. Using an electrophysiological approach in presymptomatic YAC128 HD slices, we observed decreased ascorbic acid flux from astrocytes to neurons, which altered neuronal metabolic substrate preferences. Ascorbic acid efflux and recycling were also decreased in cultured astrocytes from YAC128 HD mice. We confirmed our findings using GFAP-HD160Q, an HD mice model expressing mutant N-terminal Htt mainly in astrocytes. For the first time, we demonstrated that ascorbic acid is released from astrocytes via extracellular vesicles (EVs). Decreased number of particles and exosomal markers were observed in EV fractions from cultured YAC128 HD astrocytes and Htt-KD cells. We observed reduced number of multivesicular bodies (MVBs) in YAC128 HD striatum via electron microscopy, suggesting mutant Htt alters MVB biogenesis. EVs containing ascorbic acid effectively reduced reactive oxygen species, whereas "free" ascorbic acid played a role in modulating neuronal metabolic substrate preferences. These findings suggest that the early redox imbalance observed in HD arises from a reduced release of ascorbic acid-containing EVs by astrocytes. Meanwhile, a decrease in "free" ascorbic acid likely contributes to presymptomatic metabolic impairment.
Collapse
Affiliation(s)
- Felipe A Beltrán
- Instituto de Bioquímica y Microbiología, UACh, Valdivia, Chile; Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile
| | - Leandro Torres-Díaz L
- Instituto de Bioquímica y Microbiología, UACh, Valdivia, Chile; Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile; Departamento de Ciencias y Geografía, Facultad de Ciencias Naturales y Exactas, Universidad de Playa Ancha, Valparaíso, Chile
| | - Paulina Troncoso-Escudero
- Instituto de Bioquímica y Microbiología, UACh, Valdivia, Chile; Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile
| | - Juan Villalobos-González
- Instituto de Bioquímica y Microbiología, UACh, Valdivia, Chile; Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile
| | - Gonzalo Mayorga-Weber
- Instituto de Bioquímica y Microbiología, UACh, Valdivia, Chile; Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile
| | - Marcelo Lara
- Departamento de Biología, Universidad de Santiago de Chile, Santiago, Chile; Escuela de Química y Farmacia, Facultad de Medicina y Ciencia, Universidad San Sebastián, Campus las Tres Pascualas, Concepción, Chile
| | - Adriana Covarrubias-Pinto
- Instituto de Bioquímica y Microbiología, UACh, Valdivia, Chile; Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile; Institute of Biochemistry II, Goethe University School of Medicine, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Sharin Valdivia
- Instituto de Bioquímica y Microbiología, UACh, Valdivia, Chile; Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile; Department of Biological and Chemical Sciences, Faculty of Medicine and Sciences, San Sebastián University, Tres Pascualas Campus, Concepción, Chile
| | - Isidora Vicencio
- Instituto de Bioquímica y Microbiología, UACh, Valdivia, Chile; Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile
| | - Eduardo Papic
- Instituto de Bioquímica y Microbiología, UACh, Valdivia, Chile; Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile
| | - Carolina Paredes-Martínez
- Instituto de Bioquímica y Microbiología, UACh, Valdivia, Chile; Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile
| | - Mara E Silva-Januàrio
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Alejandro Rojas
- Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile; Instituto de Medicina, UACh, Valdivia, Chile
| | - Luis L P daSilva
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Felipe Court
- Center for Aging Research and Healthy Longevity, Faculty of Sciences, Universidad Mayor, Santiago, Chile; Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile; Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Luis Federico Bátiz
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de Los Andes, Santiago, Chile; Escuela de Medicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Patricio Rojas
- Departamento de Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Francisco J Rivera
- Translational Regenerative Neurobiology Group (TReN), Molecular and Integrative Biosciences Research Program (MIBS), Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland.
| | - Maite A Castro
- Instituto de Bioquímica y Microbiología, UACh, Valdivia, Chile; Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile; Janelia Research Campus HHMI, Ashburn, VA, USA.
| |
Collapse
|
15
|
Porel P, Bala K, Aran KR. Exploring the role of HIF-1α on pathogenesis in Alzheimer's disease and potential therapeutic approaches. Inflammopharmacology 2025; 33:669-678. [PMID: 39465478 DOI: 10.1007/s10787-024-01585-x] [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: 10/03/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024]
Abstract
Hypoxia-inducible factor 1α (HIF-1α) is a crucial transcription factor that regulates cellular responses to low oxygen levels (hypoxia). In Alzheimer's disease (AD), emerging evidence suggests a significant involvement of HIF-1α in disease pathogenesis. AD is characterized by the accumulation of amyloid-beta (Aβ) plaques and neurofibrillary tangles (NFTs), leading to neuronal dysfunction and cognitive decline. HIF-1α is implicated in AD through its multifaceted roles in various cellular processes. Firstly, in response to hypoxia, HIF-1α promotes the expression of genes involved in angiogenesis, which is crucial for maintaining cerebral blood flow and oxygen delivery to the brain. However, in the context of AD, dysregulated HIF-1α activation may exacerbate cerebral hypoperfusion, contributing to neuronal damage. Moreover, HIF-1α is implicated in the regulation of Aβ metabolism. It can influence the production and clearance of Aβ peptides, potentially modulating their accumulation and toxicity in the brain. Additionally, HIF-1α activation has been linked to neuroinflammation, a key feature of AD pathology. It can promote the expression of pro-inflammatory cytokines and exacerbate neuronal damage. Furthermore, HIF-1α may play a role in synaptic plasticity and neuronal survival, which are impaired in AD. Dysregulated HIF-1α signaling could disrupt these processes, contributing to cognitive decline and neurodegeneration. Overall, the involvement of HIF-1α in various aspects of AD pathophysiology highlights its potential as a therapeutic target. Modulating HIF-1α activity could offer novel strategies for mitigating neurodegeneration and preserving cognitive function in AD patients. However, further research is needed to elucidate the precise mechanisms underlying HIF-1α dysregulation in AD and to develop targeted interventions.
Collapse
Affiliation(s)
- Pratyush Porel
- Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Kanchan Bala
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Khadga Raj Aran
- Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India.
| |
Collapse
|
16
|
Yip JMX, Chiang GSH, Lee ICJ, Lehming-Teo R, Dai K, Dongol L, Wang LYT, Teo D, Seah GT, Lehming N. Mitochondria and the Repurposing of Diabetes Drugs for Off-Label Health Benefits. Int J Mol Sci 2025; 26:364. [PMID: 39796218 PMCID: PMC11719901 DOI: 10.3390/ijms26010364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 12/23/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
This review describes our current understanding of the role of the mitochondria in the repurposing of the anti-diabetes drugs metformin, gliclazide, GLP-1 receptor agonists, and SGLT2 inhibitors for additional clinical benefits regarding unhealthy aging, long COVID, mental neurogenerative disorders, and obesity. Metformin, the most prominent of these diabetes drugs, has been called the "Drug of Miracles and Wonders," as clinical trials have found it to be beneficial for human patients suffering from these maladies. To promote viral replication in all infected human cells, SARS-CoV-2 stimulates the infected liver cells to produce glucose and to export it into the blood stream, which can cause diabetes in long COVID patients, and metformin, which reduces the levels of glucose in the blood, was shown to cut the incidence rate of long COVID in half for all patients recovering from SARS-CoV-2. Metformin leads to the phosphorylation of the AMP-activated protein kinase AMPK, which accelerates the import of glucose into cells via the glucose transporter GLUT4 and switches the cells to the starvation mode, counteracting the virus. Diabetes drugs also stimulate the unfolded protein response and thus mitophagy, which is beneficial for healthy aging and mental health. Diabetes drugs were also found to mimic exercise and help to reduce body weight.
Collapse
Affiliation(s)
- Joyce Mei Xin Yip
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore (R.L.-T.)
| | - Grace Shu Hui Chiang
- Well Programme, Alexandra Hospital, National University Health System, Singapore 159964, Singapore; (G.S.H.C.)
| | - Ian Chong Jin Lee
- NUS High School of Mathematics and Science, Singapore 129957, Singapore
| | - Rachel Lehming-Teo
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore (R.L.-T.)
| | - Kexin Dai
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore (R.L.-T.)
| | - Lokeysh Dongol
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore (R.L.-T.)
| | - Laureen Yi-Ting Wang
- Well Programme, Alexandra Hospital, National University Health System, Singapore 159964, Singapore; (G.S.H.C.)
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore 119074, Singapore
- Division of Cardiology, Department of Medicine, Alexandra Hospital, National University Health System, Singapore 159964, Singapore
| | - Denise Teo
- Chi Longevity, Camden Medical Centre #10-04, 1 Orchard Blvd, Singapore 248649, Singapore
| | - Geok Teng Seah
- Clifford Dispensary, 77 Robinson Rd #06-02, Singapore 068896, Singapore
| | - Norbert Lehming
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore (R.L.-T.)
| |
Collapse
|
17
|
Lyamtsev AS, Sentyabreva AV, Tsvetkov IS, Miroshnichenko EA, Kosyreva AM. Morphological and Molecular Biological Changes in the Hippocampus and Prefrontal Cortex of the Brain of Newborn Male and Female Wistar Rats after LPS-Induced Activation of the Maternal Immune Response. Bull Exp Biol Med 2025; 178:381-386. [PMID: 39945955 DOI: 10.1007/s10517-025-06341-x] [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: 07/09/2024] [Indexed: 02/28/2025]
Abstract
Infectious and inflammatory processes during pregnancy in women provoke maternal immunity activation (MIA) and increase the risk of neuropsychiatric disorders in children. These disorders can lead to neurodegenerative diseases later in life. To study these effects, we evaluated the morphological and molecular biological changes in the hippocampus and prefrontal cortex of male and female Wistar rats on the 1st day of postnatal ontogenesis after LPS-induced MIA. The level of calprotectin in the blood serum of postpartum rats, the number and morphological properties of microglial cells in the hippocampus, and the expression of proinflammatory, stem, and adaptation markers in fragments of the prefrontal cortex in offspring of both sexes were determined. It was found that LPS-induced MIA had a negative effect on the developing offspring, with an increase in the level of expression of Nfκb and App in the prefrontal cortex of newborns being observed. Sex differences in morphological and molecular biological changes in the brains of newborn Wistar rats were also revealed: the number of microglial cells increased in male rats, while the number of ramified microglial cells decreased in female rats. In addition, only in females, the expression levels of the mRNA markers for stem cells, Sox2 and Sox9, decreased, while the expression level of Hif1α, which has a neuroprotective effect, increased only in males. These data may explain the differences in the incidence of neurodegenerative diseases among elderly patients of different sexes.
Collapse
Affiliation(s)
- A S Lyamtsev
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia
| | - A V Sentyabreva
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia.
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia named after Patrice Lumumba (RUDN University), Moscow, Russia.
| | - I S Tsvetkov
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia
| | - E A Miroshnichenko
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia named after Patrice Lumumba (RUDN University), Moscow, Russia
| | - A M Kosyreva
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia named after Patrice Lumumba (RUDN University), Moscow, Russia
| |
Collapse
|
18
|
Lin TK, Huang CR, Lin KJ, Hsieh YH, Chen SD, Lin YC, Chao AC, Yang DI. Potential Roles of Hypoxia-Inducible Factor-1 in Alzheimer's Disease: Beneficial or Detrimental? Antioxidants (Basel) 2024; 13:1378. [PMID: 39594520 PMCID: PMC11591038 DOI: 10.3390/antiox13111378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
The major pathological characteristics of Alzheimer's disease (AD) include senile plaques and neurofibrillary tangles (NFTs), which are mainly composed of aggregated amyloid-beta (Aβ) peptide and hyperphosphorylated tau protein, respectively. The excessive production of reactive oxygen species (ROS) and neuroinflammation are crucial contributing factors to the pathological mechanisms of AD. Hypoxia-inducible factor-1 (HIF-1) is a transcription factor critical for tissue adaption to low-oxygen tension. Growing evidence has suggested HIF-1 as a potential therapeutic target for AD; conversely, other experimental findings indicate that HIF-1 induction contributes to AD pathogenesis. These previous findings thus point to the complex, even contradictory, roles of HIF-1 in AD. In this review, we first introduce the general pathogenic mechanisms of AD as well as the potential pathophysiological roles of HIF-1 in cancer, immunity, and oxidative stress. Based on current experimental evidence in the literature, we then discuss the possible beneficial as well as detrimental mechanisms of HIF-1 in AD; these sections also include the summaries of multiple chemical reagents and proteins that have been shown to exert beneficial effects in AD via either the induction or inhibition of HIF-1.
Collapse
Affiliation(s)
- Tsu-Kung Lin
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; (T.-K.L.); (C.-R.H.); (S.-D.C.)
- College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan
| | - Chi-Ren Huang
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; (T.-K.L.); (C.-R.H.); (S.-D.C.)
- College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan
| | - Kai-Jung Lin
- Department of Family Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan;
| | - Yi-Heng Hsieh
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
| | - Shang-Der Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; (T.-K.L.); (C.-R.H.); (S.-D.C.)
| | - Yi-Chun Lin
- Department of Neurology, Taipei City Hospital Renai Branch, Taipei 106243, Taiwan;
| | - A-Ching Chao
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
- Department of Neurology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Department of Sports Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Ding-I Yang
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| |
Collapse
|
19
|
Mahanta AK, Chaulagain B, Trivedi R, Singh J. Mannose-Functionalized Chitosan-Coated PLGA Nanoparticles for Brain-Targeted Codelivery of CBD and BDNF for the Treatment of Alzheimer's Disease. ACS Chem Neurosci 2024; 15:4021-4032. [PMID: 39377785 PMCID: PMC11870748 DOI: 10.1021/acschemneuro.4c00392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024] Open
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease causing cognitive and memory decline. AD is characterized by the deposition of amyloid-β and hypophosphorylated forms of tau protein. AD brains are found to be associated with neurodegeneration, oxidative stress, and inflammation. Cannabidiol (CBD) shows neuroprotective, antioxidant, and anti-inflammatory properties and simultaneously reduces amyloid-β production and tau hyperphosphorylation. The brain-derived neurotrophic factor (BDNF) plays a vital role in the development and maintenance of the plasticity of the central nervous system. A decline of BDNF levels in AD patients results in reduced plasticity and neuronal cell death. Current therapeutics against AD are limited to only symptomatic relief, necessitating a therapeutic strategy that reverses cognitive decline. In this scenario, combination therapy of CBD and BDNF could be a fruitful strategy for the treatment of AD. We designed mannose-conjugated chitosan-coated poly(d,l-lactide-co-glycolide (PLGA) (CHTMAN-PLGA) nanoparticles for the codelivery of CBD and BDNF to the brain. Chitosan is modified with mannose to specifically target the glucose transporter-1 (GLUT-1) receptor abundantly present in the blood-brain barrier for selectively delivering therapeutics to the brain. The CBD-encapsulated nanoparticles showed an average hydrodynamic diameter of 306 ± 8.12 nm and a zeta potential of 31.7 ± 1.53 mV. The coated nanoparticles prolonged encapsulated CBD release from the PLGA matrix. The coated nanoparticles exhibited sustained release of CBD for up to 22 days with 91.68 ± 2.91% release of the encapsulated drug. The coated nanoparticles, which had a high positive zeta potential (31.7 ± 1.53 mV), encapsulated the plasmid DNA. The qualitative transfection efficiency was investigated using CHTMAN-PLGA-CBD/pGFP in bEND.3, primary astrocytes, and primary neurons, while the quantitative transfection efficiency of the delivery system was determined using CHTMAN-PLGA-CBD/pBDNF. In vitro, the pBDNF transfection study revealed that the BDNF expression was 4-fold higher for CHTMAN-PLGA-CBD/pBDNF than for naked pBDNF in all of the cell lines. The cytotoxicity and hemocompatibility of the designed nanoparticles were tested in bEND.3 cells and red blood cells, respectively, and the nanoparticles were found to be nontoxic and hemocompatible. Hence, mannose-conjugated chitosan-coated PLGA nanoparticles could be useful as brain-targeting delivery vehicles for the codelivery of CBD and BDNF for possible AD treatment.
Collapse
Affiliation(s)
- Arun Kumar Mahanta
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota 58108-6050, United States
| | - Bivek Chaulagain
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota 58108-6050, United States
| | - Riddhi Trivedi
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota 58108-6050, United States
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota 58108-6050, United States
| |
Collapse
|
20
|
Mishra S, Stany B, Das A, Kanagavel D, Vijayan M. A Comprehensive Review of Membrane Transporters and MicroRNA Regulation in Alzheimer's Disease. Mol Neurobiol 2024; 61:8739-8758. [PMID: 38558361 DOI: 10.1007/s12035-024-04135-2] [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: 11/22/2023] [Accepted: 03/15/2024] [Indexed: 04/04/2024]
Abstract
Alzheimer's disease (AD) is a distressing neurodegenerative condition characterized by the accumulation of amyloid-beta (Aβ) plaques and tau tangles within the brain. The interconnectedness between membrane transporters (SLCs) and microRNAs (miRNAs) in AD pathogenesis has gained increasing attention. This review explores the localization, substrates, and functions of SLC transporters in the brain, emphasizing the roles of transporters for glutamate, glucose, nucleosides, and other essential compounds. The examination delves into the significance of SLCs in AD, their potential for drug development, and the intricate realm of miRNAs, encompassing their transcription, processing, functions, and regulation. MiRNAs have emerged as significant players in AD, including those associated with mitochondria and synapses. Furthermore, this review discusses the intriguing nexus of miRNAs targeting SLC transporters and their potential as therapeutic targets in AD. Finally, the review underscores the interaction between SLC transporters and miRNA regulation within the context of Alzheimer's disease, underscoring the need for further research in this area. This comprehensive review aims to shed light on the complex mechanisms underlying the causation of AD and provides insights into potential therapeutic approaches.
Collapse
Affiliation(s)
- Shatakshi Mishra
- School of Biosciences and Technology, Department of Biotechnology, VIT University, Vellore, Tamil Nadu, 632014, India
| | - B Stany
- School of Biosciences and Technology, Department of Biotechnology, VIT University, Vellore, Tamil Nadu, 632014, India
| | - Anushka Das
- School of Biosciences and Technology, Department of Biotechnology, VIT University, Vellore, Tamil Nadu, 632014, India
| | - Deepankumar Kanagavel
- School of Biosciences and Technology, Department of Biotechnology, VIT University, Vellore, Tamil Nadu, 632014, India.
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX, 79430, USA.
| |
Collapse
|
21
|
Wang MY, Zhou Y, Li WL, Zhu LQ, Liu D. Friend or foe: Lactate in neurodegenerative diseases. Ageing Res Rev 2024; 101:102452. [PMID: 39127445 DOI: 10.1016/j.arr.2024.102452] [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: 06/18/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
Lactate, a byproduct of glycolysis, was considered as a metabolic waste until identified by studies on the Warburg effect. Increasing evidence elucidates that lactate functions as energy fuel, signaling molecule, and donor for protein lactylation. Altered lactate utilization is a common metabolic feature of the onset and progression of neurodegenerative diseases, such as Alzheimer's disease, multiple sclerosis, amyotrophic lateral sclerosis, Parkinson's disease and Huntington's disease. This review offers an overview of lactate metabolism from the perspective of production, transportation and clearance, and the role of lactate in neurodegenerative progression, as well as a summary of protein lactylation and the signaling function of lactate in neurodegenerative diseases. Besides, this review delves into the dual roles of changed lactate metabolism during neurodegeneration and explores prospective therapeutic methods targeting lactate. We propose that elucidating the correlation between lactate and neurodegeneration is pivotal for exploring innovative therapeutic interventions for neurodegenerative diseases.
Collapse
Affiliation(s)
- Ming-Yu Wang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yang Zhou
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wen-Lian Li
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Dan Liu
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
22
|
Méndez-Flores OG, Hernández-Kelly LC, Olivares-Bañuelos TN, López-Ramírez G, Ortega A. Brain energetics and glucose transport in metabolic diseases: role in neurodegeneration. Nutr Neurosci 2024; 27:1199-1210. [PMID: 38294500 DOI: 10.1080/1028415x.2024.2306427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
OBJECTIVES Neurons and glial cells are the main functional and structural elements of the brain, and the former depends on the latter for their nutritional, functional and structural organization, as well as for their energy maintenance. METHODS Glucose is the main metabolic source that fulfills energetic demands, either by direct anaplerosis or through its conversion to metabolic intermediates. Development of some neurodegenerative diseases have been related with modifications in the expression and/or function of glial glucose transporters, which might cause physiological and/or pathological disturbances of brain metabolism. In the present contribution, we summarized the experimental findings that describe the exquisite adjustment in expression and function of glial glucose transporters from physiologic to pathologic metabolism, and its relevance to neurodegenerative diseases. RESULTS A exhaustive literature review was done in order to gain insight into the role of brain energetics in neurodegenerative disease. This study made evident a critical involvement of glucose transporters and thus brain energetics in the development of neurodegenerative diseases. DISCUSSION An exquisite adjustment in the expression and function of glial glucose transporters from physiologic to pathologic metabolism is a biochemical signature of neurodegenerative diseases.
Collapse
Affiliation(s)
- Orquídea G Méndez-Flores
- División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco (UJAT), Villahermosa, México
| | - Luisa C Hernández-Kelly
- Laboratorio de Neurotoxicología, Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, México
| | | | - Gabriel López-Ramírez
- División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco (UJAT), Villahermosa, México
| | - Arturo Ortega
- Laboratorio de Neurotoxicología, Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, México
| |
Collapse
|
23
|
Kale MB, Bhondge HM, Wankhede NL, Shende PV, Thanekaer RP, Aglawe MM, Rahangdale SR, Taksande BG, Pandit SB, Upaganlawar AB, Umekar MJ, Kopalli SR, Koppula S. Navigating the intersection: Diabetes and Alzheimer's intertwined relationship. Ageing Res Rev 2024; 100:102415. [PMID: 39002642 DOI: 10.1016/j.arr.2024.102415] [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: 06/14/2024] [Revised: 07/06/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024]
Abstract
Alzheimer's disease (AD) and Diabetes mellitus (DM) exhibit comparable pathophysiological pathways. Genetic abnormalities in APP, PS-1, and PS-2 are linked to AD, with diagnostic aid from CSF and blood biomarkers. Insulin dysfunction, termed "type 3 diabetes mellitus" in AD, involves altered insulin signalling and neuronal shrinkage. Insulin influences beta-amyloid metabolism, exacerbating neurotoxicity in AD and amyloid production in DM. Both disorders display impaired glucose transporter expression, hastening cognitive decline. Mitochondrial dysfunction and Toll-like receptor 4-mediated inflammation worsen neurodegeneration in both diseases. ApoE4 raises disease risk, especially when coupled with dyslipidemia common in DM. Targeting shared pathways like insulin-degrading enzyme activation and HSP60 holds promise for therapeutic intervention. Recognizing these interconnected mechanisms underscores the imperative for developing tailored treatments addressing the overlapping pathophysiology of AD and DM, offering potential avenues for more effective management of both conditions.
Collapse
Affiliation(s)
- Mayur B Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | | | - Nitu L Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Prajwali V Shende
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Rushikesh P Thanekaer
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Manish M Aglawe
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Sandip R Rahangdale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Brijesh G Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Sunil B Pandit
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India
| | - Aman B Upaganlawar
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India
| | - Milind J Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| |
Collapse
|
24
|
Sultana OF, Bandaru M, Islam MA, Reddy PH. Unraveling the complexity of human brain: Structure, function in healthy and disease states. Ageing Res Rev 2024; 100:102414. [PMID: 39002647 PMCID: PMC11384519 DOI: 10.1016/j.arr.2024.102414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/29/2024] [Accepted: 07/05/2024] [Indexed: 07/15/2024]
Abstract
The human brain stands as an intricate organ, embodying a nexus of structure, function, development, and diversity. This review delves into the multifaceted landscape of the brain, spanning its anatomical intricacies, diverse functional capacities, dynamic developmental trajectories, and inherent variability across individuals. The dynamic process of brain development, from early embryonic stages to adulthood, highlights the nuanced changes that occur throughout the lifespan. The brain, a remarkably complex organ, is composed of various anatomical regions, each contributing uniquely to its overall functionality. Through an exploration of neuroanatomy, neurophysiology, and electrophysiology, this review elucidates how different brain structures interact to support a wide array of cognitive processes, sensory perception, motor control, and emotional regulation. Moreover, it addresses the impact of age, sex, and ethnic background on brain structure and function, and gender differences profoundly influence the onset, progression, and manifestation of brain disorders shaped by genetic, hormonal, environmental, and social factors. Delving into the complexities of the human brain, it investigates how variations in anatomical configuration correspond to diverse functional capacities across individuals. Furthermore, it examines the impact of neurodegenerative diseases on the structural and functional integrity of the brain. Specifically, our article explores the pathological processes underlying neurodegenerative diseases, such as Alzheimer's, Parkinson's, and Huntington's diseases, shedding light on the structural alterations and functional impairments that accompany these conditions. We will also explore the current research trends in neurodegenerative diseases and identify the existing gaps in the literature. Overall, this article deepens our understanding of the fundamental principles governing brain structure and function and paves the way for a deeper understanding of individual differences and tailored approaches in neuroscience and clinical practice-additionally, a comprehensive understanding of structural and functional changes that manifest in neurodegenerative diseases.
Collapse
Affiliation(s)
- Omme Fatema Sultana
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Madhuri Bandaru
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Md Ariful Islam
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA 5. Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
25
|
Di Domenico F, Lanzillotta C, Perluigi M. Redox imbalance and metabolic defects in the context of Alzheimer disease. FEBS Lett 2024; 598:2047-2066. [PMID: 38472147 DOI: 10.1002/1873-3468.14840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/12/2024] [Accepted: 02/12/2024] [Indexed: 03/14/2024]
Abstract
Redox reactions play a critical role for intracellular processes, including pathways involved in metabolism and signaling. Reactive oxygen species (ROS) act either as second messengers or generators of protein modifications, fundamental mechanisms for signal transduction. Disturbance of redox homeostasis is associated with many disorders. Among these, Alzheimer's disease is a neurodegenerative pathology that presents hallmarks of oxidative damage such as increased ROS production, decreased activity of antioxidant enzymes, oxidative modifications of macromolecules, and changes in mitochondrial homeostasis. Interestingly, alteration of redox homeostasis is closely associated with defects of energy metabolism, involving both carbohydrates and lipids, the major energy fuels for the cell. As the brain relies exclusively on glucose metabolism, defects of glucose utilization represent a harmful event for the brain. During aging, a progressive perturbation of energy metabolism occurs resulting in brain hypometabolism. This condition contributes to increase neuronal cell vulnerability ultimately resulting in cognitive impairment. The current review discusses the crosstalk between alteration of redox homeostasis and brain energy defects that seems to act in concert in promoting Alzheimer's neurodegeneration.
Collapse
Affiliation(s)
- Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Italy
| | - Chiara Lanzillotta
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Italy
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Italy
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
26
|
Affuso F, Micillo F, Fazio S. Insulin Resistance, a Risk Factor for Alzheimer's Disease: Pathological Mechanisms and a New Proposal for a Preventive Therapeutic Approach. Biomedicines 2024; 12:1888. [PMID: 39200352 PMCID: PMC11351221 DOI: 10.3390/biomedicines12081888] [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: 06/12/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Peripheral insulin resistance (IR) is a well-documented, independent risk factor for the development of type 2 diabetes, cardiovascular disease, cancer and cellular senescence. Recently, the brain has also been identified as an insulin-responsive region, where insulin acts as regulator of the brain metabolism. Despite the clear link between IR and the brain, the exact mechanisms underlying this relationship remain unclear. Therapeutic intervention in patients showing symptoms of neurodegenerative diseases has produced little or no results. It has been demonstrated that insulin resistance plays a significant role in the pathogenesis of neurodegenerative diseases, particularly cognitive decline. Peripheral and brain IR may represent a modifiable state that could be used to prevent major brain disorders. In this review, we will analyse the scientific literature supporting IR as a risk factor for Alzheimer's disease and suggest some therapeutic strategies to provide a new proposal for the prevention of brain IR and its consequences.
Collapse
Affiliation(s)
- Flora Affuso
- Independent Researcher, Viale Raffaello, 74, 80129 Napoli, Italy
| | - Filomena Micillo
- UOC of Geriatric Medicine AORN S.G. Moscati, 83100 Avellino, Italy
| | - Serafino Fazio
- Department of Internal Medicine, School of Medicine, Federico II University of Naples, 80138 Naples, Italy;
| |
Collapse
|
27
|
Leclerc M, Tremblay C, Bourassa P, Schneider JA, Bennett DA, Calon F. Lower GLUT1 and unchanged MCT1 in Alzheimer's disease cerebrovasculature. J Cereb Blood Flow Metab 2024; 44:1417-1432. [PMID: 38441044 PMCID: PMC11342728 DOI: 10.1177/0271678x241237484] [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: 05/03/2023] [Revised: 12/21/2023] [Accepted: 01/16/2024] [Indexed: 03/06/2024]
Abstract
The brain is a highly demanding organ, utilizing mainly glucose but also ketone bodies as sources of energy. Glucose transporter-1 (GLUT1) and monocarboxylates transporter-1 (MCT1) respectively transport glucose and ketone bodies across the blood-brain barrier. While reduced glucose uptake by the brain is one of the earliest signs of Alzheimer's disease (AD), no change in the uptake of ketone bodies has been evidenced yet. To probe for changes in GLUT1 and MCT1, we performed Western immunoblotting in microvessel extracts from the parietal cortex of 60 participants of the Religious Orders Study. Participants clinically diagnosed with AD had lower cerebrovascular levels of GLUT1, whereas MCT1 remained unchanged. GLUT1 reduction was associated with lower cognitive scores. No such association was found for MCT1. GLUT1 was inversely correlated with neuritic plaques and cerebrovascular β-secretase-derived fragment levels. No other significant associations were found between both transporters, markers of Aβ and tau pathologies, sex, age at death or apolipoprotein-ε4 genotype. These results suggest that, while a deficit of GLUT1 may underlie the reduced transport of glucose to the brain in AD, no such impairment occurs for MCT1. This study thus supports the exploration of ketone bodies as an alternative energy source for the aging brain.
Collapse
Affiliation(s)
- Manon Leclerc
- Faculté de pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec – Université Laval, Québec, Canada
| | - Cyntia Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec – Université Laval, Québec, Canada
| | - Philippe Bourassa
- Faculté de pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec – Université Laval, Québec, Canada
| | - Julie A Schneider
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Frédéric Calon
- Faculté de pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec – Université Laval, Québec, Canada
| |
Collapse
|
28
|
Daida T, Shin BC, Cepeda C, Devaskar SU. Neurodevelopment Is Dependent on Maternal Diet: Placenta and Brain Glucose Transporters GLUT1 and GLUT3. Nutrients 2024; 16:2363. [PMID: 39064806 PMCID: PMC11279700 DOI: 10.3390/nu16142363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/09/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Glucose is the primary energy source for most mammalian cells and its transport is affected by a family of facilitative glucose transporters (GLUTs) encoded by the SLC2 gene. GLUT1 and GLUT3, highly expressed isoforms in the blood-brain barrier and neuronal membranes, respectively, are associated with multiple neurodevelopmental disorders including epilepsy, dyslexia, ADHD, and autism spectrum disorder (ASD). Dietary therapies, such as the ketogenic diet, are widely accepted treatments for patients with the GLUT1 deficiency syndrome, while ameliorating certain symptoms associated with GLUT3 deficiency in animal models. A ketogenic diet, high-fat diet, and calorie/energy restriction during prenatal and postnatal stages can also alter the placental and brain GLUTs expression with long-term consequences on neurobehavior. This review focuses primarily on the role of diet/energy perturbations upon GLUT isoform-mediated emergence of neurodevelopmental and neurodegenerative disorders.
Collapse
Affiliation(s)
- Tomoko Daida
- Department of Pediatrics, Division of Neonatology and Developmental Biology and Neonatal Research Center, at the UCLA Children’s Discovery and Innovation Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.D.); (B.-C.S.)
| | - Bo-Chul Shin
- Department of Pediatrics, Division of Neonatology and Developmental Biology and Neonatal Research Center, at the UCLA Children’s Discovery and Innovation Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.D.); (B.-C.S.)
| | - Carlos Cepeda
- Intellectual and Developmental Disabilities Research Center and Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Sherin U. Devaskar
- Department of Pediatrics, Division of Neonatology and Developmental Biology and Neonatal Research Center, at the UCLA Children’s Discovery and Innovation Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.D.); (B.-C.S.)
| |
Collapse
|
29
|
Tao B, Gong W, Xu C, Ma Z, Mei J, Chen M. The relationship between hypoxia and Alzheimer's disease: an updated review. Front Aging Neurosci 2024; 16:1402774. [PMID: 39086755 PMCID: PMC11288848 DOI: 10.3389/fnagi.2024.1402774] [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: 03/18/2024] [Accepted: 07/04/2024] [Indexed: 08/02/2024] Open
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases, and the most prevalent form of dementia. The main hallmarks for the diagnosis of AD are extracellular amyloid-beta (Aβ) plaque deposition and intracellular accumulation of highly hyperphosphorylated Tau protein as neurofibrillary tangles. The brain consumes more oxygen than any other organs, so it is more easily to be affected by hypoxia. Hypoxia has long been recognized as one of the possible causes of AD and other neurodegenerative diseases, but the exact mechanism has not been clarified. In this review, we will elucidate the connection between hypoxia-inducible factors-1α and AD, including its contribution to AD and its possible protective effects. Additionally, we will discuss the relationship between oxidative stress and AD as evidence show that oxidative stress acts on AD-related pathogenic factors such as mitochondrial dysfunction, Aβ deposition, inflammation, etc. Currently, there is no cure for AD. Given the close association between hypoxia, oxidative stress, and AD, along with current research on the protective effects of antioxidants against AD, we speculate that antioxidants could be a potential therapeutic approach for AD and worth further study.
Collapse
Affiliation(s)
- Borui Tao
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- The First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Wei Gong
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chengyuan Xu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Zhihui Ma
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jinyu Mei
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ming Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
30
|
Rogal J, Zamproni LN, Nikolakopoulou P, Ygberg S, Wedell A, Wredenberg A, Herland A. Human In Vitro Models of Neuroenergetics and Neurometabolic Disturbances: Current Advances and Clinical Perspectives. Stem Cells Transl Med 2024; 13:505-514. [PMID: 38588471 PMCID: PMC11165162 DOI: 10.1093/stcltm/szae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/23/2024] [Indexed: 04/10/2024] Open
Abstract
Neurological conditions conquer the world; they are the leading cause of disability and the second leading cause of death worldwide, and they appear all around the world in every age group, gender, nationality, and socioeconomic class. Despite the growing evidence of an immense impact of perturbations in neuroenergetics on overall brain function, only little is known about the underlying mechanisms. Especially human insights are sparse, owing to a shortage of physiologically relevant model systems. With this perspective, we aim to explore the key steps and considerations involved in developing an advanced human in vitro model for studying neuroenergetics. We discuss biological and technological strategies to meet the requirements of a predictive model, aiming at providing a guide and inspiration for future in vitro models of neuroenergetics.
Collapse
Affiliation(s)
- Julia Rogal
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
- Division of Nanobiotechnology, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology at Science for Life Laboratory, 17165 Solna, Sweden
- Center for the Advancement of Integrated Medical and Engineering Sciences (AIMES), Karolinska Institute and KTH Royal Institute of Technology, 17177 Stockholm, Sweden
| | - Laura Nicoleti Zamproni
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
- Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04039-032, Brazil
| | - Polyxeni Nikolakopoulou
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
- Center for the Advancement of Integrated Medical and Engineering Sciences (AIMES), Karolinska Institute and KTH Royal Institute of Technology, 17177 Stockholm, Sweden
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Sofia Ygberg
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, 17177 Stockholm, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17177 Stockholm, Sweden
- Neuropediatric Unit, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - Anna Wedell
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, 17177 Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institute, 17177 Stockholm, Sweden
| | - Anna Wredenberg
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, 17177 Stockholm, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17177 Stockholm, Sweden
| | - Anna Herland
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
- Division of Nanobiotechnology, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology at Science for Life Laboratory, 17165 Solna, Sweden
- Center for the Advancement of Integrated Medical and Engineering Sciences (AIMES), Karolinska Institute and KTH Royal Institute of Technology, 17177 Stockholm, Sweden
| |
Collapse
|
31
|
Mitroshina EV, Vedunova MV. The Role of Oxygen Homeostasis and the HIF-1 Factor in the Development of Neurodegeneration. Int J Mol Sci 2024; 25:4581. [PMID: 38731800 PMCID: PMC11083463 DOI: 10.3390/ijms25094581] [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: 03/13/2024] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 05/13/2024] Open
Abstract
Understanding the molecular underpinnings of neurodegeneration processes is a pressing challenge for medicine and neurobiology. Alzheimer's disease (AD) and Parkinson's disease (PD) represent the most prevalent forms of neurodegeneration. To date, a substantial body of experimental evidence has strongly implicated hypoxia in the pathogenesis of numerous neurological disorders, including AD, PD, and other age-related neurodegenerative conditions. Hypoxia-inducible factor (HIF) is a transcription factor that triggers a cell survival program in conditions of oxygen deprivation. The involvement of HIF-1α in neurodegenerative processes presents a complex and sometimes contradictory picture. This review aims to elucidate the current understanding of the interplay between hypoxia and the development of AD and PD, assess the involvement of HIF-1 in their pathogenesis, and summarize promising therapeutic approaches centered on modulating the activity of the HIF-1 complex.
Collapse
Affiliation(s)
- Elena V. Mitroshina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia;
| | | |
Collapse
|
32
|
Puris E, Saveleva L, Auriola S, Gynther M, Kanninen KM, Fricker G. Sex-specific changes in protein expression of membrane transporters in the brain cortex of 5xFAD mouse model of Alzheimer's disease. Front Pharmacol 2024; 15:1365051. [PMID: 38572427 PMCID: PMC10989684 DOI: 10.3389/fphar.2024.1365051] [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: 01/03/2024] [Accepted: 02/27/2024] [Indexed: 04/05/2024] Open
Abstract
Membrane transporters playing an important role in the passage of drugs, metabolites and nutrients across the membranes of the brain cells have been shown to be involved in pathogenesis of Alzheimer's disease (AD). However, little is known about sex-specific changes in transporter protein expression at the brain in AD. Here, we investigated sex-specific alterations in protein expression of three ATP-binding cassette (ABC) and five solute carriers (SLC) transporters in the prefrontal cortex of a commonly used model of familial AD (FAD), 5xFAD mice. Sensitive liquid chromatography tandem mass spectrometry-based quantitative targeted absolute proteomic analysis was applied for absolute quantification of transporter protein expression. We compared the changes in transporter protein expressions in 7-month-old male and female 5xFAD mice versus sex-matched wild-type mice. The study revealed a significant sex-specific increase in protein expression of ABCC1 (p = 0.007) only in male 5xFAD mice as compared to sex-matched wild-type animals. In addition, the increased protein expression of glucose transporter 1 (p = 0.01), 4F2 cell-surface antigen heavy chain (p = 0.01) and long-chain fatty acid transport protein 1 (p = 0.02) were found only in female 5xFAD mice as compared to sex-matched wild-type animals. Finally, protein expression of alanine/serine/cysteine/threonine transporter 1 was upregulated in both male (p = 0.02) and female (p = 0.002) 5xFAD mice. The study provides important information about sex-specific changes in brain cortical transporter expression in 5xFAD mice, which will facilitate drug development of therapeutic strategies for AD targeting these transporters and drug delivery research.
Collapse
Affiliation(s)
- Elena Puris
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Heidelberg, Germany
| | - Liudmila Saveleva
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Seppo Auriola
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Mikko Gynther
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Katja M. Kanninen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Heidelberg, Germany
| |
Collapse
|
33
|
Hai Y, Ren K, Zhang Y, Yang L, Cao H, Yuan X, Su L, Li H, Feng X, Liu D. HIF-1α serves as a co-linker between AD and T2DM. Biomed Pharmacother 2024; 171:116158. [PMID: 38242039 DOI: 10.1016/j.biopha.2024.116158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 12/27/2023] [Accepted: 01/11/2024] [Indexed: 01/21/2024] Open
Abstract
Alzheimer's disease (AD)-related brain deterioration is linked to the type 2 diabetes mellitus (T2DM) features hyperglycemia, hyperinsulinemia, and insulin resistance. Hypoxia as a common risk factor for both AD and T2DM. Hypoxia-inducible factor-1 alpha (HIF-1α) acts as the main regulator of the hypoxia response and may be a key target in the comorbidity of AD and T2DM. HIF-1α expression is closely related to hyperglycemia, insulin resistance, and inflammation. Tissue oxygen consumption disrupts HIF-1α homeostasis, leading to increased reactive oxygen species levels and the inhibition of insulin receptor pathway activity, causing neuroinflammation, insulin resistance, abnormal Aβ deposition, and tau hyperphosphorylation. HIF-1α activation also leads to the deposition of Aβ by promoting the abnormal shearing of amyloid precursor protein and inhibiting the degradation of Aβ, and it promotes tau hyperphosphorylation by activating oxidative stress and the activation of astrocytes, which further exasperates AD. Therefore, we believe that HIF-α has great potential as a target for the treatment of AD. Importantly, the intracellular homeostasis of HIF-1α is a more crucial factor than its expression level.
Collapse
Affiliation(s)
- Yang Hai
- Scientific Research and Experimental Center, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China; Key Laboratory of Dunhuang Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China.
| | - Ke Ren
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Yarong Zhang
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Lili Yang
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Haoshi Cao
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Xianxia Yuan
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Linling Su
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Hailong Li
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Xiaoli Feng
- Scientific Research and Experimental Center, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China; Key Laboratory of Dunhuang Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Dongling Liu
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China; Northwest Collaborative Innovation Center for Traditional Chinese Medicine, Lanzhou 730000, Gansu Province, PR China; Gansu Pharmaceutical Industry Innovation Research Institute, Lanzhou 730000, Gansu Province, PR China.
| |
Collapse
|
34
|
Reiss AB, Gulkarov S, Jacob B, Srivastava A, Pinkhasov A, Gomolin IH, Stecker MM, Wisniewski T, De Leon J. Mitochondria in Alzheimer's Disease Pathogenesis. Life (Basel) 2024; 14:196. [PMID: 38398707 PMCID: PMC10890468 DOI: 10.3390/life14020196] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive and incurable neurodegenerative disorder that primarily affects persons aged 65 years and above. It causes dementia with memory loss and deterioration in thinking and language skills. AD is characterized by specific pathology resulting from the accumulation in the brain of extracellular plaques of amyloid-β and intracellular tangles of phosphorylated tau. The importance of mitochondrial dysfunction in AD pathogenesis, while previously underrecognized, is now more and more appreciated. Mitochondria are an essential organelle involved in cellular bioenergetics and signaling pathways. Mitochondrial processes crucial for synaptic activity such as mitophagy, mitochondrial trafficking, mitochondrial fission, and mitochondrial fusion are dysregulated in the AD brain. Excess fission and fragmentation yield mitochondria with low energy production. Reduced glucose metabolism is also observed in the AD brain with a hypometabolic state, particularly in the temporo-parietal brain regions. This review addresses the multiple ways in which abnormal mitochondrial structure and function contribute to AD. Disruption of the electron transport chain and ATP production are particularly neurotoxic because brain cells have disproportionately high energy demands. In addition, oxidative stress, which is extremely damaging to nerve cells, rises dramatically with mitochondrial dyshomeostasis. Restoring mitochondrial health may be a viable approach to AD treatment.
Collapse
Affiliation(s)
- Allison B. Reiss
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Shelly Gulkarov
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Benna Jacob
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Ankita Srivastava
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Aaron Pinkhasov
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Irving H. Gomolin
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Mark M. Stecker
- The Fresno Institute of Neuroscience, Fresno, CA 93730, USA;
| | - Thomas Wisniewski
- Center for Cognitive Neurology, Departments of Neurology, Pathology and Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA;
| | - Joshua De Leon
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| |
Collapse
|
35
|
Yonamine CY, Michalani MLE, Moreira RJ, Machado UF. Glucose Transport and Utilization in the Hippocampus: From Neurophysiology to Diabetes-Related Development of Dementia. Int J Mol Sci 2023; 24:16480. [PMID: 38003671 PMCID: PMC10671460 DOI: 10.3390/ijms242216480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/12/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
The association of diabetes with cognitive dysfunction has at least 60 years of history, which started with the observation that children with type 1 diabetes mellitus (T1D), who had recurrent episodes of hypoglycemia and consequently low glucose supply to the brain, showed a deficit of cognitive capacity. Later, the growing incidence of type 2 diabetes mellitus (T2D) and dementia in aged populations revealed their high association, in which a reduced neuronal glucose supply has also been considered as a key mechanism, despite hyperglycemia. Here, we discuss the role of glucose in neuronal functioning/preservation, and how peripheral blood glucose accesses the neuronal intracellular compartment, including the exquisite glucose flux across the blood-brain barrier (BBB) and the complex network of glucose transporters, in dementia-related areas such as the hippocampus. In addition, insulin resistance-induced abnormalities in the hippocampus of obese/T2D patients, such as inflammatory stress, oxidative stress, and mitochondrial stress, increased generation of advanced glycated end products and BBB dysfunction, as well as their association with dementia/Alzheimer's disease, are addressed. Finally, we discuss how these abnormalities are accompained by the reduction in the expression and translocation of the high capacity insulin-sensitive glucose transporter GLUT4 in hippocampal neurons, which leads to neurocytoglycopenia and eventually to cognitive dysfunction. This knowledge should further encourage investigations into the beneficial effects of promising therapeutic approaches which could improve central insulin sensitivity and GLUT4 expression, to fight diabetes-related cognitive dysfunctions.
Collapse
Affiliation(s)
- Caio Yogi Yonamine
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark;
| | - Maria Luiza Estimo Michalani
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil; (M.L.E.M.); (R.J.M.)
| | - Rafael Junges Moreira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil; (M.L.E.M.); (R.J.M.)
| | - Ubiratan Fabres Machado
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil; (M.L.E.M.); (R.J.M.)
| |
Collapse
|
36
|
Su Z, Zhang G, Li X, Zhang H. Inverse correlation between Alzheimer's disease and cancer from the perspective of hypoxia. Neurobiol Aging 2023; 131:59-73. [PMID: 37572528 DOI: 10.1016/j.neurobiolaging.2023.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 06/02/2023] [Accepted: 07/03/2023] [Indexed: 08/14/2023]
Abstract
Sporadic Alzheimer's disease and cancer remain epidemiologically inversely related, and exploring the reverse pathogenesis is important for our understanding of both. Cognitive dysfunctions in Alzheimer's disease (AD) might result from the depletion of adaptive reserves in the brain. Energy storage in the brain is limited and is dynamically regulated by neurovascular and neurometabolic coupling. The research on neurodegenerative diseases has been dominated by the neurocentric view that neuronal defects cause the diseases. However, the proposal of the 2-hit vascular hypothesis in AD led us to focus on alterations in the vasculature, especially hypoperfusion. Chronic hypoxia is a feature shared by AD and cancer. It is interesting how contradicting chronic hypoxia's effects on both cancer and AD are. In this article, we discuss the potential links between the 2 diseases' etiology, from comparable upstream circumstances to diametrically opposed downstream effects. We suggest opposing potential mechanisms, including upregulation and downregulation of hypoxia-inducible factor-1α, the Warburg and reverse-Warburg effects, lactate-mediated intracellular acidic and alkaline conditions, and VDAC1-mediated apoptosis and antiapoptosis, and search for regulators that may be identified as the crossroads between cancer and AD.
Collapse
Affiliation(s)
- Zhan Su
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, China
| | - Guimei Zhang
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, China
| | - Xiangting Li
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, China
| | - Haining Zhang
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
37
|
Choi YS, Song JE, Kim E, Kim CH, Lee JE, Song HT. Hyperpolarized [1- 13C]pyruvate Magnetic Resonance Spectroscopy Shows That Agmatine Increased Lactate Production in the Brain of Type 2 Diabetic Mice. Yonsei Med J 2023; 64:625-632. [PMID: 37727922 PMCID: PMC10522877 DOI: 10.3349/ymj.2022.0554] [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: 12/23/2022] [Revised: 07/19/2023] [Accepted: 07/27/2023] [Indexed: 09/21/2023] Open
Abstract
PURPOSE Type 2 diabetes mellitus (T2DM) is associated with a 2-fold increased risk of developing Alzheimer's disease. In earlier research, agmatine has been demonstrated to alleviate diabetes symptoms and increase cognitive performance. However, it is unclear whether the improvement of cognitive function is attributable to the reduction of diabetic symptoms or its direct influence on brain metabolism. Using hyperpolarized (HP) [1-13C]pyruvate magnetic resonance spectroscopy (MRS), this study intends to evaluate the influence of agmatine on brain metabolism. MATERIALS AND METHODS ICR mice were fed a high-fat diet and injected with streptozotocin to develop a T2DM animal model. During a 2-week period, T2DM mice were treated with normal saline or 100 mg/kg of agmatine, and brain HP [1-13C]pyruvate MRS was performed. The effect of agmatine on lactate generation and NADH/NAD+ redox state was investigated using C6 and neuro-2a (N2a) cells. RESULTS As a perfusion marker, the total 13C signals in the brain of T2DM mice (p=0.07) and agmatine-treated mice (p<0.05) were reduced. The conversion constant (Kpl) from [1-13C]pyruvate to [1-13C]lactate was not distinguishable in the brains of T2DM mice but was significantly increased in the brains of agmatine-treated T2DM mice. Treating C6 and N2a cells with agmatine increased NADH/NAD+ratio and lactate generation. CONCLUSION Agmatine influences the NADH/NAD+ redox state in the brains of T2DM mice, which may be connected with enhanced cognitive performance and increased conversion of HP [1-13C]pyruvate to HP [1-13C]lactate.
Collapse
Affiliation(s)
- Young-Suk Choi
- Department of Radiology and Research Institute of Radiological Science, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Eun Song
- Department of Radiology, Stanford University, Palo Alto, CA, USA
- Promaxo Inc., Oakland, CA, USA
| | - Eosu Kim
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Chul Hoon Kim
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea
- BK21 PLUS Project for Medical Sciences and Brain Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jong Eun Lee
- BK21 PLUS Project for Medical Sciences and Brain Research Institute, Yonsei University College of Medicine, Seoul, Korea
- Department of Anatomy and Research Institute of Radiological Science, Yonsei University College of Medicine, Seoul, Korea.
| | - Ho-Taek Song
- Department of Radiology and Research Institute of Radiological Science, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
38
|
Raut S, Bhalerao A, Powers M, Gonzalez M, Mancuso S, Cucullo L. Hypometabolism, Alzheimer's Disease, and Possible Therapeutic Targets: An Overview. Cells 2023; 12:2019. [PMID: 37626828 PMCID: PMC10453773 DOI: 10.3390/cells12162019] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/19/2023] [Accepted: 08/06/2023] [Indexed: 08/27/2023] Open
Abstract
The brain is a highly dynamic organ that requires a constant energy source to function normally. This energy is mostly supplied by glucose, a simple sugar that serves as the brain's principal fuel source. Glucose transport across the blood-brain barrier (BBB) is primarily controlled via sodium-independent facilitated glucose transport, such as by glucose transporter 1 (GLUT1) and 3 (GLUT3). However, other glucose transporters, including GLUT4 and the sodium-dependent transporters SGLT1 and SGLT6, have been reported in vitro and in vivo. When the BBB endothelial layer is crossed, neurons and astrocytes can absorb the glucose using their GLUT1 and GLUT3 transporters. Glucose then enters the glycolytic pathway and is metabolized into adenosine triphosphate (ATP), which supplies the energy to support cellular functions. The transport and metabolism of glucose in the brain are impacted by several medical conditions, which can cause neurological and neuropsychiatric symptoms. Alzheimer's disease (AD), Parkinson's disease (PD), epilepsy, traumatic brain injury (TBI), schizophrenia, etc., are a few of the most prevalent disorders, characterized by a decline in brain metabolism or hypometabolism early in the course of the disease. Indeed, AD is considered a metabolic disorder related to decreased brain glucose metabolism, involving brain insulin resistance and age-dependent mitochondrial dysfunction. Although the conventional view is that reduced cerebral metabolism is an effect of neuronal loss and consequent brain atrophy, a growing body of evidence points to the opposite, where hypometabolism is prodromal or at least precedes the onset of brain atrophy and the manifestation of clinical symptoms. The underlying processes responsible for these glucose transport and metabolic abnormalities are complicated and remain poorly understood. This review article provides a comprehensive overview of the current understanding of hypometabolism in AD and potential therapeutic targets.
Collapse
Affiliation(s)
- Snehal Raut
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA; (S.R.); (A.B.); (M.G.); (S.M.)
| | - Aditya Bhalerao
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA; (S.R.); (A.B.); (M.G.); (S.M.)
| | - Michael Powers
- Department of Biological and Biomedical Sciences, Oakland University, Rochester, MI 48309, USA;
| | - Minelly Gonzalez
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA; (S.R.); (A.B.); (M.G.); (S.M.)
| | - Salvatore Mancuso
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA; (S.R.); (A.B.); (M.G.); (S.M.)
| | - Luca Cucullo
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA; (S.R.); (A.B.); (M.G.); (S.M.)
| |
Collapse
|
39
|
Samman WA, Selim SM, El Fayoumi HM, El-Sayed NM, Mehanna ET, Hazem RM. Dapagliflozin Ameliorates Cognitive Impairment in Aluminum-Chloride-Induced Alzheimer's Disease via Modulation of AMPK/mTOR, Oxidative Stress and Glucose Metabolism. Pharmaceuticals (Basel) 2023; 16:ph16050753. [PMID: 37242536 DOI: 10.3390/ph16050753] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/02/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurological illness characterized by memory loss and cognitive deterioration. Dapagliflozin was suggested to attenuate the memory impairment associated with AD; however, its mechanisms were not fully elucidated. This study aims to examine the possible mechanisms of the neuroprotective effects of dapagliflozin against aluminum chloride (AlCl3)-induced AD. Rats were distributed into four groups: group 1 received saline, group 2 received AlCl3 (70 mg/kg) daily for 9 weeks, and groups 3 and 4 were administered AlCl3 (70 mg/kg) daily for 5 weeks. Dapagliflozin (1 mg/kg) and dapagliflozin (5 mg/kg) were then given daily with AlCl3 for another 4 weeks. Two behavioral experiments were performed: the Morris Water Maze (MWM) and the Y-maze spontaneous alternation (Y-maze) task. Histopathological alterations in the brain, as well as changes in acetylcholinesterase (AChE) and amyloid β (Aβ) peptide activities and oxidative stress (OS) markers, were all evaluated. A western blot analysis was used for the detection of phosphorylated 5' AMP-activated protein kinase (p-AMPK), phosphorylated mammalian target of Rapamycin (p-mTOR) and heme oxygenase-1 (HO-1). Tissue samples were collected for the isolation of glucose transporters (GLUTs) and glycolytic enzymes using PCR analysis, and brain glucose levels were also measured. The current data demonstrate that dapagliflozin represents a possible approach to combat AlCl3-induced AD in rats through inhibiting oxidative stress, enhancing glucose metabolism and activating AMPK signaling.
Collapse
Affiliation(s)
- Waad A Samman
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Medina 30078, Saudi Arabia
| | - Salma M Selim
- Department of Pharmacology and Toxicology, Faculty of Dentistry, Sinai University, Kantara, Ismailia 41636, Egypt
| | - Hassan M El Fayoumi
- Department of Pharmacology and Toxicology, Faculty of Dentistry, Sinai University, Kantara, Ismailia 41636, Egypt
| | - Norhan M El-Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Eman T Mehanna
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Reem M Hazem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
40
|
Sung JH, Ou Y, Barger SW. Amyloid β-Peptide Effects on Glucose Regulation Are Dependent on Apolipoprotein E Genotype. eNeuro 2023; 10:ENEURO.0376-22.2023. [PMID: 37163733 PMCID: PMC10135078 DOI: 10.1523/eneuro.0376-22.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 01/26/2023] [Accepted: 02/14/2023] [Indexed: 05/12/2023] Open
Abstract
The apolipoprotein E gene (APOE) confers the greatest genetic risk factor for Alzheimer's disease (AD), wherein the ε4 allele confers an elevated risk compared with the ε3 allele. Biological mechanisms that differ across these alleles have been explored in mouse models wherein the murine Apoe gene has undergone targeted replacement with sequences encoding human ApoE3 or ApoE4 (ApoE-TR mice). Such models have indicated that the two variants of ApoE produce differential effects on energy metabolism, including metabolic syndrome. However, glucose regulation has not been compared in ApoE-TR mice with and without amyloid β-peptide (Aβ) accumulation. We crossed ApoE3-TR and ApoE4-TR mice with a transgenic line that accumulates human Aβ1-42 In male ApoE3-TR mice, introduction of Aβ caused aberrations in glucose tolerance and in membrane translocation of astrocytic glucose transporter 1 (GLUT1). Phosphorylation of Tau at AD-relevant sites was correlated with glucose intolerance. These effects appeared independent of insulin dysregulation and were not observed in females. In ApoE4-TR mice, the addition of Aβ had no significant effects because of a trend toward perturbation of the baseline values.
Collapse
Affiliation(s)
- Jin Hee Sung
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205
| | - Yang Ou
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205
| | - Steven W Barger
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205
- Geriatric Research, Education and Clinical Center, Central Arkansas Veterans Healthcare System, Little Rock, AR 72205
| |
Collapse
|
41
|
Yang YY, Ren YT, Jia MY, Bai CY, Liang XT, Gao HL, Zhong ML, Wang T, Guo C. The human islet amyloid polypeptide reduces hippocampal tauopathy and behavioral impairments in P301S mice without inducing neurotoxicity or seeding amyloid aggregation. Exp Neurol 2023; 362:114346. [PMID: 36750170 DOI: 10.1016/j.expneurol.2023.114346] [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: 10/26/2022] [Revised: 01/17/2023] [Accepted: 01/30/2023] [Indexed: 02/07/2023]
Abstract
Recent evidence suggests that human islet amyloid polypeptide (h-IAPP) accumulates in the brains of Alzheimer's disease (AD) patients and may interact with Aβ or microtubule associated protein tau to associate with the neurodegenerative process. Increasing evidence indicates a potential protective effect of h-IAPP against Aβ-induced neurotoxicity in AD mouse models. However, a direct therapeutic effect of h-IAPP supplementation on tauopathy has not been established. Here, we found that long-term h-IAPP treatment attenuated tau hyperphosphorylation levels and induced neuroinflammation and oxidative damage, prevented synaptic loss and neuronal degeneration in the hippocampus, and alleviated behavioral deficits in P301S transgenic mice (a mouse model of tauopathy). Restoration of insulin sensitization, glucose/energy metabolism, and activated BDNF signaling also contributed to the underlying mechanisms. These findings suggest that seemly h-IAPP has promise for the treatment of neurodegenerative disorders with tauopathy, such as AD.
Collapse
Affiliation(s)
- Ying-Ying Yang
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China; Liaoning Cheng Da Biotechnology Co., Ltd, Shenyang 110179, China
| | - Yan-Tao Ren
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China
| | - Meng-Yu Jia
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China; Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China
| | - Chen-Yang Bai
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China; Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China
| | - Xiu-Ting Liang
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China; Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China
| | - Hui-Ling Gao
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China
| | - Man-Li Zhong
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China
| | - Tao Wang
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China
| | - Chuang Guo
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China; Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China.
| |
Collapse
|
42
|
The Involvement of Post-Translational Modifications in Regulating the Development and Progression of Alzheimer's Disease. Mol Neurobiol 2023; 60:3617-3632. [PMID: 36877359 DOI: 10.1007/s12035-023-03277-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 02/16/2023] [Indexed: 03/07/2023]
Abstract
Post-translational modifications (PTMs) have been recently reported to be involved in the development and progression of Alzheimer's disease (AD). In detail, PTMs include phosphorylation, glycation, acetylation, sumoylation, ubiquitination, methylation, nitration, and truncation, which are associated with pathological functions of AD-related proteins, such as β-amyloid (Aβ), β-site APP-cleavage enzyme 1 (BACE1), and tau protein. In particular, the roles of aberrant PTMs in the trafficking, cleavage, and degradation of AD-associated proteins, leading to the cognitive decline of the disease, are summarized under AD conditions. By summarizing these research progress, the gaps will be filled between PMTs and AD, which will facilitate the discovery of potential biomarkers, leading to the establishment of novel clinical intervention methods against AD.
Collapse
|
43
|
Zhang Y, Geng R, Liu M, Deng S, Ding J, Zhong H, Tu Q. Shared peripheral blood biomarkers for Alzheimer’s disease, major depressive disorder, and type 2 diabetes and cognitive risk factor analysis. Heliyon 2023; 9:e14653. [PMID: 36994393 PMCID: PMC10040717 DOI: 10.1016/j.heliyon.2023.e14653] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/28/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
Background Alzheimer's disease (AD), type 2 diabetes mellitus (T2DM), and Major Depressive Disorder (MDD) have a higher incidence rate in modern society. Although increasing evidence supports close associations between the three, the mechanisms underlying their interrelationships remain elucidated. Objective The primary purpose is to explore the shared pathogenesis and the potential peripheral blood biomarkers for AD, MDD, and T2DM. Methods We downloaded the microarray data of AD, MDD, and T2DM from the Gene Expression Omnibus database and constructed co-expression networks by Weighted Gene Co-Expression Network Analysis to identify differentially expressed genes. We took the intersection of differentially expressed genes to obtain co-DEGs. Then, we performed GO and KEGG enrichment analysis on the common genes in the AD, MDD, and T2DM-related modules. Next, we utilized the STRING database to find the hub genes in the protein-protein interaction network. ROC curves were constructed for co-DEGs to obtain the most diagnostic valuable genes and to make drug predictions against the target genes. Finally, we conducted a present condition survey to verify the correlation between T2DM, MDD and AD. Results Our findings indicated 127 diff co-DEGs, 19 upregulated co-DEGs, and 25 down-regulated co-DEGs. Functional enrichment analysis showed co-DEGs were mainly enriched in signaling pathways such as metabolic diseases and some neurodegeneration. Protein-protein interaction network construction identified hub genes in AD, MDD and T2DM shared genes. We identified seven hub genes of co-DEGs, namely, SMC4, CDC27, HNF1A, RHOD, CUX1, PDLIM5, and TTR. The current survey results suggest a correlation between T2DM, MDD and dementia. Moreover, logistic regression analysis showed that T2DM and depression increased the risk of dementia. Conclusion Our work identified common pathogenesis of AD, T2DM, and MDD. These shared pathways might provide novel ideas for further mechanistic studies and hub genes that may serve as novel therapeutic targets for diagnosing and treating.
Collapse
|
44
|
Yoon JH, Hwang J, Son SU, Choi J, You SW, Park H, Cha SY, Maeng S. How Can Insulin Resistance Cause Alzheimer's Disease? Int J Mol Sci 2023; 24:3506. [PMID: 36834911 PMCID: PMC9966425 DOI: 10.3390/ijms24043506] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/17/2023] [Accepted: 01/27/2023] [Indexed: 02/12/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder associated with cognitive decline. Despite worldwide efforts to find a cure, no proper treatment has been developed yet, and the only effective countermeasure is to prevent the disease progression by early diagnosis. The reason why new drug candidates fail to show therapeutic effects in clinical studies may be due to misunderstanding the cause of AD. Regarding the cause of AD, the most widely known is the amyloid cascade hypothesis, in which the deposition of amyloid beta and hyperphosphorylated tau is the cause. However, many new hypotheses were suggested. Among them, based on preclinical and clinical evidence supporting a connection between AD and diabetes, insulin resistance has been pointed out as an important factor in the development of AD. Therefore, by reviewing the pathophysiological background of brain metabolic insufficiency and insulin insufficiency leading to AD pathology, we will discuss how can insulin resistance cause AD.
Collapse
Affiliation(s)
- Ji Hye Yoon
- Age-Tech Service Convergence Major, Graduate School of East–West Medical Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
| | - JooHyun Hwang
- Age-Tech Service Convergence Major, Graduate School of East–West Medical Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
| | - Sung Un Son
- Department of Comprehensive Health Science, Graduate School of East–West Medical Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
| | - Junhyuk Choi
- Age-Tech Service Convergence Major, Graduate School of East–West Medical Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
| | - Seung-Won You
- Department of Comprehensive Health Science, Graduate School of East–West Medical Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
| | - Hyunwoo Park
- Department of Comprehensive Health Science, Graduate School of East–West Medical Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
- Health Park Co., Ltd., Seoul 02447, Republic of Korea
| | - Seung-Yun Cha
- Department of Comprehensive Health Science, Graduate School of East–West Medical Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
| | - Sungho Maeng
- Age-Tech Service Convergence Major, Graduate School of East–West Medical Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
- Department of Comprehensive Health Science, Graduate School of East–West Medical Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
| |
Collapse
|
45
|
Abstract
Probabilistic and parsimony-based arguments regarding available genetics data are used to propose that Hardy and Higgin's amyloid cascade hypothesis is valid but is commonly interpreted too narrowly to support, incorrectly, the primacy of the amyloid-β peptide (Aβ) in driving Alzheimer's disease pathogenesis. Instead, increased activity of the βCTF (C99) fragment of AβPP is the critical pathogenic determinant altered by mutations in the APP gene. This model is consistent with the regulation of APP mRNA translation via its 5' iron responsive element. Similar arguments support that the pathological effects of familial Alzheimer's disease mutations in the genes PSEN1 and PSEN2 are not exerted directly via changes in AβPP cleavage to produce different ratios of Aβ length. Rather, these mutations likely act through effects on presenilin holoprotein conformation and function, and possibly the formation and stability of multimers of presenilin holoprotein and/or of the γ-secretase complex. All fAD mutations in APP, PSEN1, and PSEN2 likely find unity of pathological mechanism in their actions on endolysosomal acidification and mitochondrial function, with detrimental effects on iron homeostasis and promotion of "pseudo-hypoxia" being of central importance. Aβ production is enhanced and distorted by oxidative stress and accumulates due to decreased lysosomal function. It may act as a disease-associated molecular pattern enhancing oxidative stress-driven neuroinflammation during the cognitive phase of the disease.
Collapse
Affiliation(s)
- Michael Lardelli
- Alzheimer's Disease Genetics Laboratory, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
46
|
Yan F, Liu J, Chen MX, Zhang Y, Wei SJ, Jin H, Nie J, Fu XL, Shi JS, Zhou SY, Jin F. Icariin ameliorates memory deficits through regulating brain insulin signaling and glucose transporters in 3ΧTg-AD mice. Neural Regen Res 2023; 18:183-188. [PMID: 35799540 PMCID: PMC9241391 DOI: 10.4103/1673-5374.344840] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
47
|
Omori NE, Malys MK, Woo G, Mansor L. Exploring the role of ketone bodies in the diagnosis and treatment of psychiatric disorders. Front Psychiatry 2023; 14:1142682. [PMID: 37139329 PMCID: PMC10149735 DOI: 10.3389/fpsyt.2023.1142682] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/28/2023] [Indexed: 05/05/2023] Open
Abstract
In recent times, advances in the field of metabolomics have shed greater light on the role of metabolic disturbances in neuropsychiatric conditions. The following review explores the role of ketone bodies and ketosis in both the diagnosis and treatment of three major psychiatric disorders: major depressive disorder, anxiety disorders, and schizophrenia. Distinction is made between the potential therapeutic effects of the ketogenic diet and exogenous ketone preparations, as exogenous ketones in particular offer a standardized, reproducible manner for inducing ketosis. Compelling associations between symptoms of mental distress and dysregulation in central nervous system ketone metabolism have been demonstrated in preclinical studies with putative neuroprotective effects of ketone bodies being elucidated, including effects on inflammasomes and the promotion of neurogenesis in the central nervous system. Despite emerging pre-clinical data, clinical research on ketone body effectiveness as a treatment option for psychiatric disorders remains lacking. This gap in understanding warrants further investigating, especially considering that safe and acceptable ways of inducing ketosis are readily available.
Collapse
Affiliation(s)
- Naomi Elyse Omori
- Health Via Modern Nutrition Inc. (H.V.M.N.), San Francisco, CA, United States
- *Correspondence: Naomi Elyse Omori,
| | - Mantas Kazimieras Malys
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College, London, United Kingdom
| | - Geoffrey Woo
- Health Via Modern Nutrition Inc. (H.V.M.N.), San Francisco, CA, United States
| | - Latt Mansor
- Health Via Modern Nutrition Inc. (H.V.M.N.), San Francisco, CA, United States
| |
Collapse
|
48
|
Bakrim S, Aboulaghras S, El Menyiy N, El Omari N, Assaggaf H, Lee LH, Montesano D, Gallo M, Zengin G, AlDhaheri Y, Bouyahya A. Phytochemical Compounds and Nanoparticles as Phytochemical Delivery Systems for Alzheimer's Disease Management. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27249043. [PMID: 36558176 PMCID: PMC9781052 DOI: 10.3390/molecules27249043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/04/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease remains one of the most widespread neurodegenerative reasons for dementia worldwide and is associated with considerable mortality and morbidity. Therefore, it has been considered a priority for research. Indeed, several risk factors are involved in the complexity of the therapeutic ways of this pathology, including age, traumatic brain injury, genetics, exposure to aluminum, infections, diabetes, vascular diseases, hypertension, dyslipidemia, and obesity. The pathophysiology of Alzheimer's disease is mostly associated with hyperphosphorylated protein in the neuronal cytoplasm and extracellular plaques of the insoluble β-amyloid peptide. Therefore, the management of this pathology needs the screening of drugs targeting different pathological levels, such as acetylcholinesterase (AchE), amyloid β formation, and lipoxygenase inhibitors. Among the pharmacological strategies used for the management of Alzheimer's disease, natural drugs are considered a promising therapeutic strategy. Indeed, bioactive compounds isolated from different natural sources exhibit important anti-Alzheimer effects by their effectiveness in promoting neuroplasticity and protecting against neurodegeneration as well as neuroinflammation and oxidative stress in the brain. These effects involve different sub-cellular, cellular, and/or molecular mechanisms, such as the inhibition of acetylcholinesterase (AchE), the modulation of signaling pathways, and the inhibition of oxidative stress. Moreover, some nanoparticles were recently used as phytochemical delivery systems to improve the effects of phytochemical compounds against Alzheimer's disease. Therefore, the present work aims to provide a comprehensive overview of the key advances concerning nano-drug delivery applications of phytochemicals for Alzheimer's disease management.
Collapse
Affiliation(s)
- Saad Bakrim
- Geo-Bio-Environment Engineering and Innovation Laboratory, Molecular Engineering, Biotechnology and Innovation Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir 80000, Morocco
| | - Sara Aboulaghras
- Physiology and Physiopathology Team, Faculty of Sciences, Genomic of Human Pathologies Research, Mohammed V University in Rabat, Rabat 10100, Morocco
| | - Naoual El Menyiy
- Laboratory of Pharmacology, National Agency of Medicinal and Aromatic Plants, Taounate 34025, Morocco
| | - Nasreddine El Omari
- Laboratory of Histology, Embryology and Cytogenetic, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat 10100, Morocco
| | - Hamza Assaggaf
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya 47500, Malaysia
| | - Domenico Montesano
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy
| | - Monica Gallo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy
| | - Gokhan Zengin
- Department of Biology, Science Faculty, Selcuk University, 42130 Konya, Turkey
- Correspondence: (G.Z.); (Y.A.); (A.B.)
| | - Yusra AlDhaheri
- Department of Biology, College of Science, United Arab Emirates University, Al Ain 15551, United Arab Emirates
- Correspondence: (G.Z.); (Y.A.); (A.B.)
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco
- Correspondence: (G.Z.); (Y.A.); (A.B.)
| |
Collapse
|
49
|
HIF-1α Causes LCMT1/PP2A Deficiency and Mediates Tau Hyperphosphorylation and Cognitive Dysfunction during Chronic Hypoxia. Int J Mol Sci 2022; 23:ijms232416140. [PMID: 36555780 PMCID: PMC9783654 DOI: 10.3390/ijms232416140] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
Chronic hypoxia is a risk factor for Alzheimer's disease (AD), and the neurofibrillary tangle (NFT) formed by hyperphosphorylated tau is one of the two major pathological changes in AD. However, the effect of chronic hypoxia on tau phosphorylation and its mechanism remains unclear. In this study, we investigated the role of HIF-1α (the functional subunit of hypoxia-inducible factor 1) in tau pathology. It was found that in Sprague-Dawley (SD) rats, global hypoxia (10% O2, 6 h per day) for one month induced cognitive impairments. Meanwhile it induced HIF-1α increase, tau hyperphosphorylation, and protein phosphatase 2A (PP2A) deficiency with leucine carboxyl methyltransferase 1(LCMT1, increasing PP2A activity) decrease in the rats' hippocampus. The results were replicated by hypoxic treatment in primary hippocampal neurons and C6/tau cells (rat C6 glioma cells stably expressing human full-length tau441). Conversely, HIF-1α silencing impeded the changes induced by hypoxia, both in primary neurons and SD rats. The result of dual luciferase assay proved that HIF-1α acted as a transcription factor of LCMT1. Unexpectedly, HIF-1α decreased the protein level of LCMT1. Further study uncovered that both overexpression of HIF-1α and hypoxia treatment resulted in a sizable degradation of LCMT1 via the autophagy--lysosomal pathway. Together, our data strongly indicated that chronic hypoxia upregulates HIF-1α, which obviously accelerated LCMT1 degradation, thus counteracting its transcriptional expression. The increase in HIF-1α decreases PP2A activity, finally resulting in tau hyperphosphorylation and cognitive dysfunction. Lowering HIF-1α in chronic hypoxia conditions may be useful in AD prevention.
Collapse
|
50
|
Weber CM, Moiz B, Zic SM, Alpízar Vargas V, Li A, Clyne AM. Induced pluripotent stem cell-derived cells model brain microvascular endothelial cell glucose metabolism. Fluids Barriers CNS 2022; 19:98. [PMID: 36494870 PMCID: PMC9733016 DOI: 10.1186/s12987-022-00395-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/27/2022] [Indexed: 12/13/2022] Open
Abstract
Glucose transport from the blood into the brain is tightly regulated by brain microvascular endothelial cells (BMEC), which also use glucose as their primary energy source. To study how BMEC glucose transport contributes to cerebral glucose hypometabolism in diseases such as Alzheimer's disease, it is essential to understand how these cells metabolize glucose. Human primary BMEC (hpBMEC) can be used for BMEC metabolism studies; however, they have poor barrier function and may not recapitulate in vivo BMEC function. iPSC-derived BMEC-like cells (hiBMEC) are readily available and have good barrier function but may have an underlying epithelial signature. In this study, we examined differences between hpBMEC and hiBMEC glucose metabolism using a combination of dynamic metabolic measurements, metabolic mass spectrometry, RNA sequencing, and Western blots. hiBMEC had decreased glycolytic flux relative to hpBMEC, and the overall metabolomes and metabolic enzyme levels were different between the two cell types. However, hpBMEC and hiBMEC had similar glucose metabolism, including nearly identical glucose labeled fractions of glycolytic and TCA cycle metabolites. Treatment with astrocyte conditioned media and high glucose increased glycolysis in both hpBMEC and hiBMEC, though hpBMEC decreased glycolysis in response to fluvastatin while hiBMEC did not. Together, these results suggest that hiBMEC can be used to model cerebral vascular glucose metabolism, which expands their use beyond barrier models.
Collapse
Affiliation(s)
| | - Bilal Moiz
- University of Maryland, College Park, MD, 20742, USA
| | - Sophia M Zic
- University of Maryland, College Park, MD, 20742, USA
| | | | - Andrew Li
- University of Maryland, College Park, MD, 20742, USA
| | | |
Collapse
|