1
|
Yadang SAF, Nguezeye Y, Taiwe GS, Agbor GA, Ngo Bum E. Therapeutic effects of Carissa edulis aqueous extract against L-glutamic acid-induced neurotoxicity in brain mice. IBRO Neurosci Rep 2025; 18:453-463. [PMID: 40162365 PMCID: PMC11951986 DOI: 10.1016/j.ibneur.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 04/02/2025] Open
Abstract
The over-stimulation of N-methyl-d-aspartate glutamate receptors causes an excitotoxic neuronal death which plays an important role in many neurodegenerative diseases. Carissa edulis, a medicinal plant used in African pharmacopeia has been shown to have many therapeutic effects. In this study, the therapeutic effects of Carissa edulis aqueous extract on L-glutamic acid-induced neurotoxicity in mice were investigated. Two-month-old mice received an intraperitoneal injection of L-glutamic acid (2 g/kg) for seven consecutive days and were treated with an aqueous extract of Carissa edulis. Mice were monitored for behavioural studies including locomotion, muscle strength, and memory. Oxidative stress was determined by measuring lipid peroxidation and the level of antioxidant enzymes. Elisa kits were used to evaluate the levels of the proinflammatory cytokines. Hippocampal histopathology was examined using cresyl violet staining. Carissa edulis administration exhibited a protective effect on L-glutamic acid-induced abnormal locomotor activity and increased the mice's muscle strength. Also, it increased the memory of treated mice. Carissa edulis aqueous extract administration decreased the malondialdehyde level and increased the catalase activity and glutathione level. Furthermore, it significantly decreased the levels of IL-1β and TNF-α compared to the L-glutamic acid group. There were no significant pathological changes in the hippocampus of the Carissa edulis-treated group compared to the L-glutamic acid group. Our results indicated that Carissa edulis aqueous extract showed therapeutic effects by alleviating memory impairment, decreasing oxidative stress, and proinflammatory cytokines in the brains of mice treated with L-glutamic acid. Therefore, Carissa edulis treatment may help reduce glutamatergic neurotoxicity in neurodegenerative diseases.
Collapse
Affiliation(s)
- Sabine Adeline Fanta Yadang
- Laboratory of Pharmacology and Drug Discovery, Centre for Research on Medicinal Plants and Traditional Medicine, Institute of Medical Research and Medicinal Plants Studies, Yaounde, Cameroon
| | - Yvette Nguezeye
- Laboratory of Pharmacology and Drug Discovery, Centre for Research on Medicinal Plants and Traditional Medicine, Institute of Medical Research and Medicinal Plants Studies, Yaounde, Cameroon
| | - Germain Sotoing Taiwe
- Department of Animal Biology and Conservation, Faculty of Science, University of Buea, Buea, Cameroon
| | - Gabriel Agbor Agbor
- Laboratory of Pharmacology and Drug Discovery, Centre for Research on Medicinal Plants and Traditional Medicine, Institute of Medical Research and Medicinal Plants Studies, Yaounde, Cameroon
| | - Elisabeth Ngo Bum
- Department of Biological Sciences, Faculty of Science, University of Maroua, Maroua, Cameroon
| |
Collapse
|
2
|
Almohmadi NH, Al-Kuraishy HM, Al-Gareeb AI, Albuhadily AK, Abdelaziz AM, Jabir MS, Alexiou A, Papadakis M, Batiha GES. Glutamatergic dysfunction in neurodegenerative diseases focusing on Parkinson's disease: Role of glutamate modulators. Brain Res Bull 2025; 225:111349. [PMID: 40252703 DOI: 10.1016/j.brainresbull.2025.111349] [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: 01/16/2025] [Revised: 04/02/2025] [Accepted: 04/15/2025] [Indexed: 04/21/2025]
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disorder resulting from the degeneration of dopamenergic neurons in the substantia nigra pars compacta (SNpc). Research has predominantly centered on understanding the dysfunction of dopaminergic neurotransmission in PD. Recently, more studies discussed the potential role of other neurotransmitters in PD neuropathology. One of the most important non-dopaminergic neurotransmitters involved in the pathogenesis of PD is glutamate, which is widely involved in glutamatergic neurotransmission in different brain regions, including SNpc. The development and progression of PD neuropathology and levodopa-induced dyskinesias (LID) are associated with glutamate neurotoxicity. Therefore, this review seeks to explore the possible involvement of glutamatergic signaling in PD development and assess the therapeutic potential of glutamate receptor antagonists in treating the disorder.
Collapse
Affiliation(s)
- Najlaa Hamed Almohmadi
- Clinical Nutrition Department, College of Applied Medical Sciences, Umm Al-Qura University, Makkah 24381, Saudi Arabia.
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, Iraq.
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, Iraq; Jabir ibn Hayyan Medical University Al-Ameer Qu, Po. Box (13), Kufa, Najaf, Iraq.
| | - Ali K Albuhadily
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, Iraq.
| | - Ahmed M Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University-Arish Branch, Arish 45511, Egypt.
| | - Majid S Jabir
- Department of Applied Science, University of Technology-Iraq, Baghdad, Iraq.
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW 2770, Australia; University Centre for Research & Development, Chandigarh University, Mohali, India; Department of Research & Development, Funogen, Athens, Greece.
| | - Marios Papadakis
- University Hospital Witten-Herdecke, University of Witten, Herdecke, Heusnerstrasse 40, Wuppertal 42283, Germany.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhur University, Damanhur, AlBeheira 22511, Egypt.
| |
Collapse
|
3
|
Cotrin JC, Dos Santos Junior GC, Cadaxo AS, Pereira JS, Spitz M, de Rosso ALZ, Veras RP, Valente AP, Pimentel MMG, Santos-Rebouças CB. Plasma and urinary metabolomic signatures differentiate genetic and idiopathic Parkinson's disease. Brain Res 2025; 1858:149625. [PMID: 40204143 DOI: 10.1016/j.brainres.2025.149625] [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: 03/11/2025] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 04/11/2025]
Abstract
Parkinson's disease (PD) is marked by alpha-synuclein accumulation and progressive dopaminergic neuron loss. Using Nuclear Magnetic Resonance (NMR)-based metabolomics, we uncovered metabolic disturbances in idiopathic PD (iPD) and PD linked to LRRK2, GBA1, and PRKN variants in a Brazilian ethnically diverse cohort, free of comorbidities, in comparison to healthy, age-matched controls. In plasma, significant PD-associated metabolites included histidine, acetate, acetoacetate, glutamine, glucose, lipids and lipoproteins, N-acetyl-glycoproteins, and sarcosine. Urine samples revealed alterations in creatine, creatinine, L-asparagine, trimethylamine, 3-beta-hydroxybutyrate, isovaleric acid, glutamine, urea, glycine, choline, arginine, and cysteine in association with PD. Notably, creatine, creatinine, acetate, glucose, and histidine showed pathway influences from LRRK2, GBA1, and PRKN variants. Enrichment analyses highlighted disruptions in glyoxylate and dicarboxylate metabolism (plasma) as well as serine, threonine, and glycine metabolism (urine). Additionally, a metabolite-gene-disease interaction network identified 15 genes associated with PD that interact with key metabolites, highlighting MAPT, SNCA, RERE, and KCNN3 as key players in both plasmaandurine. NMR in saliva samples did not show significant differences between PD groups and controls. Our findings underscore PD-associated metabolites, particularly related to arginine metabolism, the urea cycle, glutamate metabolism, glucose metabolism, and gut microbiota. These pathways and gene interactions may serve as potential biomarkers for PD diagnosis and precision medicine strategies.
Collapse
Affiliation(s)
- Juliana Cordovil Cotrin
- Human Genetics Service, Department of Genetics, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Gilson Costa Dos Santos Junior
- Laboratory of Metabolomics, Department of Genetics, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - André Simões Cadaxo
- Human Genetics Service, Department of Genetics, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Joao Santos Pereira
- Movement Disorders Clinic, Neurology Service, Pedro Ernesto University Hospital, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Mariana Spitz
- Movement Disorders Clinic, Neurology Service, Pedro Ernesto University Hospital, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Ana Lúcia Zuma de Rosso
- Department of Neurology, Hospital Universitário Clementino Fraga Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renato Peixoto Veras
- Institute of Human Aging, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Ana Paula Valente
- National Center of Nuclear Magnetic Resonance, Center for Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Márcia Mattos Gonçalves Pimentel
- Human Genetics Service, Department of Genetics, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Cíntia Barros Santos-Rebouças
- Human Genetics Service, Department of Genetics, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University, Rio de Janeiro, Brazil.
| |
Collapse
|
4
|
Krueger ME, Boles JS, Simon ZD, Alvarez SD, McFarland NR, Okun MS, Zimmermann EM, Forsmark CE, Tansey MG. Comparative analysis of Parkinson's and inflammatory bowel disease gut microbiomes reveals shared butyrate-producing bacteria depletion. NPJ Parkinsons Dis 2025; 11:50. [PMID: 40108151 PMCID: PMC11923181 DOI: 10.1038/s41531-025-00894-4] [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: 05/25/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
Epidemiological studies reveal that inflammatory bowel disease (IBD) is associated with an increased risk of Parkinson's disease (PD). Gut dysbiosis has been documented in both PD and IBD, however it is currently unknown whether gut dysbiosis underlies the epidemiological association between both diseases. To identify shared and distinct features of the PD and IBD microbiome, we recruited 54 PD, 26 IBD, and 16 healthy control individuals and performed the first joint analysis of gut metagenomes. Larger, publicly available PD and IBD metagenomic datasets were also analyzed to validate and extend our findings. Depletions in short-chain fatty acid (SCFA)-producing bacteria, including Roseburia intestinalis, Faecalibacterium prausnitzii, Anaerostipes hadrus, and Eubacterium rectale, as well depletion in SCFA-synthesis pathways were detected across PD and IBD datasets, suggesting that depletion of these microbes in IBD may influence the risk for PD development.
Collapse
Affiliation(s)
- Maeve E Krueger
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Jake Sondag Boles
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Zachary D Simon
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Stephan D Alvarez
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Nikolaus R McFarland
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
- Department of Medicine, Division of Gastroenterology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Michael S Okun
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
- Department of Medicine, Division of Gastroenterology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Ellen M Zimmermann
- Department of Medicine, Division of Gastroenterology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Christopher E Forsmark
- Department of Medicine, Division of Gastroenterology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Malú Gámez Tansey
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA.
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA.
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
- Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
5
|
Salarvandian S, Digaleh H, Khodagholi F, Javadpour P, Asadi S, Zaman AAO, Dargahi L. Harmonic activity of glutamate dehydrogenase and neuroplasticity: The impact on aging, cognitive dysfunction, and neurodegeneration. Behav Brain Res 2025; 480:115399. [PMID: 39675635 DOI: 10.1016/j.bbr.2024.115399] [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: 07/25/2024] [Revised: 11/21/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024]
Abstract
In recent years, glutamate has attracted significant attention for its roles in various brain processes. However, one of its key regulators, glutamate dehydrogenase (GDH), remains understudied despite its pivotal role in several biochemical pathways. Dysfunction or dysregulation of GDH has been implicated in aging and various neurological disorders, such as Alzheimer's disease and Parkinson's disease. In this review, the impact of GDH on aging, cognitive impairment, and neurodegenerative conditions, as exemplars of the phenomena that may affected by neuroplasticity, has been reviewed. Despite extensive research on synaptic plasticity, the precise influence of GDH on brain structure and function remains undiscovered. This review of existing literature on GDH and neuroplasticity reveals diverse and occasionally conflicting effects. Future research endeavors should aim to describe the precise mechanisms by which GDH influences neuroplasticity (eg. synaptic plasticity and neurogenesis), particularly in the context of human aging and disease progression. Studies on GDH activity have been limited by factors such as insufficient sample sizes and varying experimental conditions. Researchers should focus on investigating the molecular mechanisms by which GDH modulates neuroplasticity, utilizing various animal strains and species, ages, sexes, GDH isoforms, brain regions, and cell types. Understanding GDH's role in neuroplasticity may offer innovative therapeutic strategies for neurodegenerative and psychiatric diseases, potentially slowing the aging process and promoting brain regeneration.
Collapse
Affiliation(s)
- Shakiba Salarvandian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hadi Digaleh
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Neurosurgery, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Pegah Javadpour
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sareh Asadi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Ali Orang Zaman
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Capó T, Rebassa JB, Raïch I, Lillo J, Badia P, Navarro G, Reyes-Resina I. Future Perspectives of NMDAR in CNS Disorders. Molecules 2025; 30:877. [PMID: 40005187 PMCID: PMC11857888 DOI: 10.3390/molecules30040877] [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: 01/14/2025] [Revised: 02/05/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Neurodegenerative diseases such as Alzheimer's and Parkinson's diseases are among the leading causes of physical and cognitive disability across the globe. Fifty million people worldwide suffer these diseases, and that number is expected to rise as the population ages. Ictus is another pathology that also courses with neurodegeneration and is a leading cause of mortality and long-term disability in developed countries. Schizophrenia is not as common as other mental disorders, affecting approximately 24 million people worldwide. All these disorders have in common that still there is not an effective pharmacological treatment to cure them. The N-methyl-D-aspartate (NMDA) receptor (NMDAR) has attracted attention as a potential therapeutic target due to its important role in learning and memory and also due to its implication in excitotoxicity processes. Some drugs targeting NMDARs are already being used to treat symptoms of disorders affecting the central nervous system (CNS). Here, we aim to review the implications of NMDAR in these CNS pathologies, its role as a potential therapeutic target, and the future perspectives for developing new treatments focused on these receptors.
Collapse
Affiliation(s)
- Toni Capó
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; (T.C.); (J.B.R.); (I.R.); (P.B.)
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain;
- Institute of Neuroscience, University of Barcelona (NeuroUB), Campus Mundet, Passeig de la Vall d’Hebron171, 08035 Barcelona, Spain
| | - Joan Biel Rebassa
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; (T.C.); (J.B.R.); (I.R.); (P.B.)
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain;
- Institute of Neuroscience, University of Barcelona (NeuroUB), Campus Mundet, Passeig de la Vall d’Hebron171, 08035 Barcelona, Spain
| | - Iu Raïch
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; (T.C.); (J.B.R.); (I.R.); (P.B.)
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain;
- Institute of Neuroscience, University of Barcelona (NeuroUB), Campus Mundet, Passeig de la Vall d’Hebron171, 08035 Barcelona, Spain
| | - Jaume Lillo
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain;
- Institute of Neuroscience, University of Barcelona (NeuroUB), Campus Mundet, Passeig de la Vall d’Hebron171, 08035 Barcelona, Spain
- Department of Biochemistry and Molecular Biomedicine, School of Biology, University of Barcelona, 08028 Barcelona, Spain
| | - Pau Badia
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; (T.C.); (J.B.R.); (I.R.); (P.B.)
| | - Gemma Navarro
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; (T.C.); (J.B.R.); (I.R.); (P.B.)
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain;
- Institute of Neuroscience, University of Barcelona (NeuroUB), Campus Mundet, Passeig de la Vall d’Hebron171, 08035 Barcelona, Spain
| | - Irene Reyes-Resina
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; (T.C.); (J.B.R.); (I.R.); (P.B.)
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain;
- Institute of Neuroscience, University of Barcelona (NeuroUB), Campus Mundet, Passeig de la Vall d’Hebron171, 08035 Barcelona, Spain
| |
Collapse
|
7
|
Denaro S, D’Aprile S, Vicario N, Parenti R. Mechanistic insights into connexin-mediated neuroglia crosstalk in neurodegenerative diseases. Front Cell Neurosci 2025; 19:1532960. [PMID: 40007760 PMCID: PMC11850338 DOI: 10.3389/fncel.2025.1532960] [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: 11/22/2024] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Multiple Sclerosis (MS), and Huntington's disease (HD), although distinct in their clinical manifestations, share a common hallmark: a disrupted neuroinflammatory environment orchestrated by dysregulation of neuroglial intercellular communication. Neuroglial crosstalk is physiologically ensured by extracellular mediators and by the activity of connexins (Cxs), the forming proteins of gap junctions (Gjs) and hemichannels (HCs), which maintain intracellular and extracellular homeostasis. However, accumulating evidence suggests that Cxs can also act as pathological pore in neuroinflammatory conditions, thereby contributing to neurodegenerative phenomena such as synaptic dysfunction, oxidative stress, and ultimately cell death. This review explores mechanistic insights of Cxs-mediated intercellular communication in the progression of neurodegenerative diseases and discusses the therapeutic potential of targeting Cxs to restore cellular homeostasis.
Collapse
Affiliation(s)
| | | | | | - Rosalba Parenti
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
8
|
Ravera S, Farsetti E, Maura G, Marcoli M, Bozzo M, Cervetto C, Amaroli A. 810-nm Photobiomodulation Evokes Glutamate Release in Normal and Rotenone-Dysfunctional Cortical Nerve Terminals by Modulating Mitochondrial Energy Metabolism. Cells 2025; 14:67. [PMID: 39851493 PMCID: PMC11764165 DOI: 10.3390/cells14020067] [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: 11/25/2024] [Revised: 12/23/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025] Open
Abstract
The dysfunction of mitochondria, the primary source of cellular energy and producer of reactive oxygen species (ROS), is associated with brain aging and neurodegenerative diseases. Scientific evidence indicates that light in the visible and near-infrared spectrum can modulate mitochondrial activity, a phenomenon known in medicine as photobiomodulation therapy (PBM-t). The beneficial effects of PBM-t on dementia and neurodegeneration have been reviewed in the literature. However, the molecular mechanisms underlying these findings have yet to be fully elucidated. This study investigates the mechanism behind dose-dependent glutamate release in nerve terminals after irradiation with 810 nm, 1 W for 60 s continuous, 1 cm2, 1 W/cm2, 60 J, 60 J/cm2 (810 nm-1 W) or 810 nm, 0.1 W for 60 s continuous, 1 cm2, 0.1 W/cm2, 6 J, 6 J/cm2 (810 nm-0.1 W), focusing on mitochondrial activities. The results show that PBM modulated the mitochondrial metabolism of cortical nerve terminals and supported a power-dependent increase in oxidative phosphorylation (OxPhos) activity when stimulated with pyruvate plus malate (P/M) or succinate (succ) as respiratory substrates. The PBM-induced increase in OxPhos was sensitive to adding rotenone (Complex I inhibitor) and antimycin A (Complex III inhibitor) when synaptosomes were stimulated with P/M, but only to antimycin A when stimulated with succ. This allowed us to observe that the glutamate efflux, disrupted in the presence of rotenone, was partially restored by PBM due to the increase in the OxPhos pathway led by Complex II. This evidence suggests that PBM, acting on mitochondria, could facilitate physiological communication within the neuron-astrocyte network through vesicular glutamate release, potentially regulating healthy brain function and brain dysfunction.
Collapse
Affiliation(s)
- Silvia Ravera
- Department of Experimental Medicine, University of Genova, 16132 Genova, Italy;
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Elisa Farsetti
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, 16148 Genova, Italy;
| | - Guido Maura
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genova, 16132 Genova, Italy; (G.M.); (M.M.); (M.B.)
| | - Manuela Marcoli
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genova, 16132 Genova, Italy; (G.M.); (M.M.); (M.B.)
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), 56122 Pisa, Italy
| | - Matteo Bozzo
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genova, 16132 Genova, Italy; (G.M.); (M.M.); (M.B.)
| | - Chiara Cervetto
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, 16148 Genova, Italy;
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), 56122 Pisa, Italy
| | - Andrea Amaroli
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), 56122 Pisa, Italy
- BIO-Photonics Overarching Research Laboratory, Department of Earth, Environmental and Life Sciences (DISTAV), University of Genova, 16132 Genova, Italy
| |
Collapse
|
9
|
He M, Wollmuth LP. Regulation of NMDAR activation efficiency by environmental factors and subunit composition. J Gen Physiol 2025; 157:e202413637. [PMID: 39576244 PMCID: PMC11586625 DOI: 10.1085/jgp.202413637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/28/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024] Open
Abstract
NMDA receptors (NMDAR) convert the major excitatory neurotransmitter glutamate into a synaptic signal. A key question is how efficiently the ion channel opens in response to the rapid exposure to presynaptic glutamate release. Here, we applied glutamate to single channel outside-out patches and measured the successes of channel openings and the latency to first opening to assay the activation efficiency of NMDARs under different physiological conditions and with different human subunit compositions. For GluN1/GluN2A receptors, we find that various factors, including intracellular ATP and GTP, can enhance the efficiency of activation presumably via the intracellular C-terminal domain. Notably, an energy-based internal solution or increasing the time between applications to increase recovery time improved efficiency. However, even under these optimized conditions and with a 1-s glutamate application, there remained around 10-15% inefficiency. Channel activation became more inefficient with brief synaptic-like pulses of glutamate at 2 ms. Of the different NMDAR subunit compositions, GluN2B-containing NMDARs showed the lowest success rate and longest latency to first openings, highlighting that they display the most distinct activation mechanism. In contrast, putative triheteromeric GluN1/GluN2A/GluN2B receptors showed high activation efficiency. Despite the low open probability, NMDARs containing either GluN2C or GluN2D subunits displayed high activation efficiency, nearly comparable with that for GluN2A-containing receptors. These results highlight that activation efficiency in NMDARs can be regulated by environmental surroundings and varies across different subunits.
Collapse
Affiliation(s)
- Miaomiao He
- Graduate Program in Biochemistry and Structural Biology, Stony Brook University, Stony Brook, NY, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY, USA
| | - Lonnie P. Wollmuth
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
10
|
Pekdemir B, Raposo A, Saraiva A, Lima MJ, Alsharari ZD, BinMowyna MN, Karav S. Mechanisms and Potential Benefits of Neuroprotective Agents in Neurological Health. Nutrients 2024; 16:4368. [PMID: 39770989 PMCID: PMC11677798 DOI: 10.3390/nu16244368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/12/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
The brain contains many interconnected and complex cellular and molecular mechanisms. Injury to the brain causes permanent dysfunctions in these mechanisms. So, it continues to be an area where surgical intervention cannot be performed except for the removal of tumors and the repair of some aneurysms. Some agents that can cross the blood-brain barrier and reach neurons show neuroprotective effects in the brain due to their anti-apoptotic, anti-inflammatory and antioxidant properties. In particular, some agents act by reducing or modulating the accumulation of protein aggregates in neurodegenerative diseases (Alzheimer's disease, Parkinson's disease, Huntington's disease, Amyotrophic lateral sclerosis, and prion disease) caused by protein accumulation. Substrate accumulation causes increased oxidative stress and stimulates the brain's immune cells, microglia, and astrocytes, to secrete proinflammatory cytokines. Long-term or chronic neuroinflammatory response triggers apoptosis. Brain damage is observed with neuronal apoptosis and brain functions are impaired. This situation negatively affects processes such as motor movements, memory, perception, and learning. Neuroprotective agents prevent apoptosis by modulating molecules that play a role in apoptosis. In addition, they can improve impaired brain functions by supporting neuroplasticity and neurogenesis. Due to the important roles that these agents play in central nervous system damage or neurodegenerative diseases, it is important to elucidate many mechanisms. This review provides an overview of the mechanisms of flavonoids, which constitute a large part of the agents with neuroprotective effects, as well as vitamins, neurotransmitters, hormones, amino acids, and their derivatives. It is thought that understanding these mechanisms will enable the development of new therapeutic agents and different treatment strategies.
Collapse
Affiliation(s)
- Burcu Pekdemir
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey;
| | - António Raposo
- CBIOS (Research Center for Biosciences and Health Technologies), Universidade Lusófona de Humanidades e Tecnologias, Campo Grande 376, 1749-024 Lisboa, Portugal
| | - Ariana Saraiva
- Research in Veterinary Medicine (I-MVET), Faculty of Veterinary Medicine, Lisbon University Centre, Lusófona University, Campo Grande 376, 1749-024 Lisboa, Portugal;
| | - Maria João Lima
- CERNAS Research Centre, Polytechnic University of Viseu, 3504-510 Viseu, Portugal;
| | - Zayed D. Alsharari
- Department of Clinical Nutrition, Faculty of Applied Medical Sciences, University of Tabuk, P.O. Box 741, Tabuk 71491, Saudi Arabia;
| | - Mona N. BinMowyna
- College of Education, Shaqra University, Shaqra 11911, Saudi Arabia;
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey;
| |
Collapse
|
11
|
Shukla A, Meena K, Gupta A, Sandhir R. 1H NMR-Based Metabolomic Signatures in Rodent Models of Sporadic Alzheimer's Disease and Metabolic Disorders. ACS Chem Neurosci 2024; 15:4478-4499. [PMID: 39629865 DOI: 10.1021/acschemneuro.4c00510] [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: 12/19/2024] Open
Abstract
Alzheimer's disease (AD) is a chronic neurological disorder that impacts the elderly population all over the globe. Evidence suggests association between AD and metabolic disorders such as diabetes mellitus (DM) and obesity (OB). The present study is an attempt to evaluate metabolic alterations in the serum and brain through NMR spectroscopy with the aim to identify shared metabolic signatures. AD was induced in rats by stereotactic intracerebroventricular injection of oligomerized Aβ-42 peptide into the brain. DM and OB were induced by intraperitoneal injection of streptozotocin and feeding rats on a high-fat diet, respectively. The metabolic alterations obtained through 1H NMR spectroscopy were further subjected to multivariate analysis by principal component analysis and partial least-squares discrimination for identification of metabolic signatures. In the serum, the levels of lactate and betaine were increased in AD, DM, and OB rats. On the other hand, the metabolite profile of brain indicated increase in the levels of lactate, N-acetylaspartate, and creatinine in AD, DM, and OB rats. Additionally, the concentration of neurochemicals such as glutamate, GABA, N-acetylglutamate, and myo-inositol were also elevated. The alterations in neurotransmitters and cerebral energy metabolism were accompanied by deficits in cognition assessed by Morris water maze in AD, DM, and OB rats. The perturbed metabolic profiles were accompanied by the presence of pathogenic amyloid deposits visualized by Congo red stain in the brains of AD, DM, and OB rats. Overall, the study identifies common metabolic signatures in AD, DM, and OB that may be involved in etiopathogenesis and also suggests linkages between these three conditions.
Collapse
Affiliation(s)
- Ananya Shukla
- Department of Biochemistry, Hargobind Khorana Block (BMS Block II), Panjab University, Sector-25, Chandigarh 160014, India
| | - Khushbhu Meena
- Centre of Bio-Medical Research, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS) Campus, Lucknow, Uttar Pradesh 226014, India
| | - Ashish Gupta
- Centre of Bio-Medical Research, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS) Campus, Lucknow, Uttar Pradesh 226014, India
| | - Rajat Sandhir
- Department of Biochemistry, Hargobind Khorana Block (BMS Block II), Panjab University, Sector-25, Chandigarh 160014, India
| |
Collapse
|
12
|
Domin H, Burnat G. mGlu4R, mGlu7R, and mGlu8R allosteric modulation for treating acute and chronic neurodegenerative disorders. Pharmacol Rep 2024; 76:1219-1241. [PMID: 39348087 PMCID: PMC11582148 DOI: 10.1007/s43440-024-00657-7] [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: 07/19/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 10/01/2024]
Abstract
Neuroprotection, defined as safeguarding neurons from damage and death by inhibiting diverse pathological mechanisms, continues to be a promising approach for managing a range of central nervous system (CNS) disorders, including acute conditions such as ischemic stroke and traumatic brain injury (TBI) and chronic neurodegenerative diseases like Parkinson's disease (PD), Alzheimer's disease (AD), and multiple sclerosis (MS). These pathophysiological conditions involve excessive glutamatergic (Glu) transmission activity, which can lead to excitotoxicity. Inhibiting this excessive Glu transmission has been proposed as a potential therapeutic strategy for treating the CNS disorders mentioned. In particular, ligands of G protein-coupled receptors (GPCRs), including metabotropic glutamatergic receptors (mGluRs), have been recognized as promising options for inhibiting excessive Glu transmission. This review discusses the complex interactions of mGlu receptors with their subtypes, including the formation of homo- and heterodimers, which may vary in function and pharmacology depending on their protomer composition. Understanding these intricate details of mGlu receptor structure and function enhances researchers' ability to develop targeted pharmacological interventions, potentially offering new therapeutic avenues for neurological and psychiatric disorders. This review also summarizes the current knowledge of the neuroprotective potential of ligands targeting group III mGluRs in preclinical cellular (in vitro) and animal (in vivo) models of ischemic stroke, TBI, PD, AD, and MS. In recent years, experiments have shown that compounds, especially those activating mGlu4 or mGlu7 receptors, exhibit protective effects in experimental ischemia models. The discovery of allosteric ligands for specific mGluR subtypes has led to reports suggesting that group III mGluRs may be promising targets for neuroprotective therapy in PD (mGlu4R), TBI (mGlu7R), and MS (mGlu8R).
Collapse
Affiliation(s)
- Helena Domin
- Maj Institute of Pharmacology, Department of Neurobiology, Polish Academy of Sciences, Smętna 12, Kraków, 31-343, Poland.
| | - Grzegorz Burnat
- Maj Institute of Pharmacology, Department of Neurobiology, Polish Academy of Sciences, Smętna 12, Kraków, 31-343, Poland
| |
Collapse
|
13
|
Liu R, Liu N, Ma L, Liu Y, Huang Z, Peng X, Zhuang C, Niu J, Yu J, Du J. Research Progress on NMDA Receptor Enhancement Drugs for the Treatment of Depressive Disorder. CNS Drugs 2024; 38:985-1002. [PMID: 39379772 DOI: 10.1007/s40263-024-01123-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2024] [Indexed: 10/10/2024]
Abstract
Major depressive disorder (MDD) is a severe mental illness with a complex etiology. Currently, many medications employed in clinical treatment exhibit limitations such as delayed onset of action and a high incidence of adverse reactions. Therefore, there is a pressing need to develop antidepressants that exhibit enhanced efficacy and safety. The N-methyl-D-aspartate receptor (NMDAR), a distinctive glutamate-gated ion channel receptor, has been implicated in the onset and progression of depressive disorder, as evidenced by both preclinical and clinical research. The NMDAR antagonist, ketamine, exhibits rapid and sustained antidepressant effects, holding promise as a novel therapeutic approach for depressive disorder. However, its psychotomimetic impact and potential for addiction have restricted its widespread clinical application. Notably, over the past decade, studies have suggested that enhancing NMDAR functionality can produce antidepressant effects with improved safety, especially with the emergence of NMDAR-positive allosteric modulators (PAMs). We view this as a potential novel strategy for treating depression, forming the basis for the narrative review that follows.
Collapse
Affiliation(s)
- Ruyun Liu
- School of Pharmacy, Ningxia Medical University, 1160 Shengli St, Xingqing District, Yinchuan, Ningxia, China
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, 1160 Shengli St, Xingqing District, Yinchuan, Ningxia, China
| | - Ning Liu
- School of Pharmacy, Ningxia Medical University, 1160 Shengli St, Xingqing District, Yinchuan, Ningxia, China
| | - Lin Ma
- School of Pharmacy, Ningxia Medical University, 1160 Shengli St, Xingqing District, Yinchuan, Ningxia, China
| | - Yue Liu
- School of Pharmacy, Ningxia Medical University, 1160 Shengli St, Xingqing District, Yinchuan, Ningxia, China
| | - Zhuo Huang
- Department of Molecular and Cellular Pharmacology, State Key Laboratory of Natural and Biomimetic Drugs, Peking University School of Pharmaceutical Sciences, Beijing, China
| | - Xiaodong Peng
- School of Pharmacy, Ningxia Medical University, 1160 Shengli St, Xingqing District, Yinchuan, Ningxia, China
| | - Chunlin Zhuang
- School of Pharmacy, Ningxia Medical University, 1160 Shengli St, Xingqing District, Yinchuan, Ningxia, China
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Jianguo Niu
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, 1160 Shengli St, Xingqing District, Yinchuan, Ningxia, China.
- School of Basic Medicine, Ningxia Medical University, 1160 Shengli St, Xingqing District, Yinchuan, Ningxia, China.
| | - Jianqiang Yu
- School of Pharmacy, Ningxia Medical University, 1160 Shengli St, Xingqing District, Yinchuan, Ningxia, China.
| | - Juan Du
- School of Pharmacy, Ningxia Medical University, 1160 Shengli St, Xingqing District, Yinchuan, Ningxia, China.
| |
Collapse
|
14
|
Wang X, Ma W, Wang Y, Ren F, Wang K, Li N. Norlignans and Phenolics from Curculigo capitulata and Their Neuroprotection Against Glutamate-Induced Oxidative Injury in SH-SY5Y Cells. Molecules 2024; 29:5648. [PMID: 39683807 DOI: 10.3390/molecules29235648] [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: 11/09/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
The herb Curculigo capitulata (Lour.) Ktze is widely distributed in southern and southwestern China. The Curculigo genus and its primary chemical constituents exhibit remarkable antidepressant activities. To investigate the chemical constituents and potential health benefits of C. capitulata, a phytochemical study was conducted. In this study, seven new compounds (capitugenin A-G), including three new norlignans (1-3), a new chalcone dimer (4), a new hemiacetal (5), two novel pyrrolidine-based compounds (6 and 7), including one identified as a natural product (7), and nineteen known compounds (8-26), were isolated from C. capitulata. The chemical structures and absolute configurations of Compounds 1-7 were elucidated via comprehensive spectroscopic data analyses. The neuroprotective effects of Compounds 1-26 against glutamate-induced cell death were tested in the human neuroblastoma cell line SH-SY5Y. Compounds 1, 3, 6, 8, 11, and 17 showed significant neuroprotective effects, with protection rates ranging from 29.4 to 52.8% at concentrations ranging from 5 to 40 μM. Western blot analysis indicated that Compound 3 exerted a protective effect by regulating the expression of Nrf2/HO-1.
Collapse
Affiliation(s)
- Xueru Wang
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Wei Ma
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Ying Wang
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Fucai Ren
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Kaijin Wang
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Ning Li
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
15
|
Nguyen DL, Le MPT, Lee KW, Kim JH, Yoon HC, Pham HTM. Development of a Disease Modeling Framework for Glutamatergic Neurons Derived from Neuroblastoma Cells in 3D Microarrays. Sci Rep 2024; 14:29144. [PMID: 39587250 PMCID: PMC11589682 DOI: 10.1038/s41598-024-80369-3] [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: 08/18/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024] Open
Abstract
Neurodegenerative diseases (NDDs) present significant challenges due to limited treatment options, ethical concerns surrounding traditional animal models, and the time-consuming and costly process of using human-induced pluripotent stem cells (iPSCs). We addressed these issues by developing a 3D culture protocol for differentiating SH-SY5Y cells into glutamatergic neurons, enhancing physiological relevance with a 3D microarray culture plate. Our protocol optimized serum concentration and incorporated retinoic acid (RA) to improve differentiation. We analyzed the proportions of N-type and S-type cells, observing that RA in the maturation stage not only reduced cell proliferation but also enhanced the expression of MAP2 and VGLUT1, indicating effective neuronal differentiation. Our approach demonstrates the strong expression of glutamatergic neuron phenotypes in 3D SH-SY5Y neural spheroids, offering a promising tool for high-throughput NDD modeling and advancing drug discovery and therapeutic development. This method overcomes limitations associated with conventional 2D cultures and animal models, providing a more effective platform for NDD research.
Collapse
Affiliation(s)
- Duc Long Nguyen
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - My Phuong Thi Le
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Kyung Won Lee
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
- Advanced College of Bio-convergence Engineering, Ajou University, Suwon, 16499, South Korea
| | - Jae-Ho Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
- ANK corporation, TheANK, Suwon, 16522, South Korea
| | - Hyun C Yoon
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea.
- Advanced College of Bio-convergence Engineering, Ajou University, Suwon, 16499, South Korea.
| | - Huyen T M Pham
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea.
| |
Collapse
|
16
|
Fujii R, Nambu Y, Sawant Shirikant N, Furube E, Morita M, Yoshimura R, Miyata S. Neuronal regeneration in the area postrema of adult mouse medulla oblongata following glutamate-induced neuronal elimination. Neuroscience 2024; 563:188-201. [PMID: 39521321 DOI: 10.1016/j.neuroscience.2024.11.009] [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: 11/26/2023] [Revised: 11/02/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Neural stem cells and/or progenitor cells (NSCs/NPCs) in the subventricular and subgranular zones of the adult mammal forebrain generate new neurons and are involved in partial repair after injury. Recently, NSCs/NPCs were identified in the area postrema (AP) of the medulla oblongata of the hindbrain. In this study, we used the properties of fenestrate capillaries to observe specific neuronal elimination in the AP of adult mice and investigated subsequent neuronal regeneration by neurogenesis. Subcutaneous administration of monosodium glutamate (MSG) induced prominent Fos expression in HuC/D+ neurons in the AP 2 h after administration. MSG administration caused a marked decrease in HuC/D+ neuronal density by neuronal death 3 to 21 days after administration, which recovered to the control level 35 days later. After MSG administration, the density of TUNEL+ dying neurons and phagocytic microglia surrounding or engulfing neurons increased. Within 7 days of MSG administration, the number of BrdU+ Sox2+ and BrdU+ Math1+ cells increased markedly, and at least the BrdU+ Math1+ cells similarly increased for the next following 7 days. A remarkable number of HuC/D+ neurons with BrdU+ nuclei were observed 35 days after MSG administration. This study reveals that neurogenesis occurs in the AP of adult mice, recovering and maintaining normal neuronal density after neuronal death.
Collapse
Affiliation(s)
- Rena Fujii
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Yuri Nambu
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Nitin Sawant Shirikant
- Department of Biology, Graduate School of Science, Kobe University, Kobe 657-8501, Japan
| | - Eriko Furube
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan; Department of Anatomy, Asahikawa Medical University School of Medicine, Midorigaoka, Asahikawa, Hokkaido 078-8510, Japan
| | - Mitsuhiro Morita
- Department of Biology, Graduate School of Science, Kobe University, Kobe 657-8501, Japan
| | - Ryoichi Yoshimura
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Seiji Miyata
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan.
| |
Collapse
|
17
|
Wang X, Rusinova R, Gregorio GG, Boudker O. Free fatty acids inhibit an ion-coupled membrane transporter by dissipating the ion gradient. J Biol Chem 2024; 300:107955. [PMID: 39491650 DOI: 10.1016/j.jbc.2024.107955] [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: 06/24/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024] Open
Abstract
Glutamate is the main excitatory transmitter in the mammalian central nervous system; glutamate transporters keep the synaptic glutamate concentrations at bay for normal brain function. Arachidonic acid (AA), docosahexaenoic acid, and other unsaturated fatty acids modulate glutamate transporters in cell- and tissue slices-based studies. Here, we investigated their effect and mechanism using a purified archaeal glutamate transporter homolog reconstituted into the lipid membranes. AA, docosahexaenoic acid, and related fatty acids irreversibly inhibited the sodium-dependent concentrative substrate uptake into lipid vesicles within the physiologically relevant concentration range. In contrast, AA did not inhibit amino acid exchange across the membrane. The length and unsaturation of the aliphatic tail affect inhibition, and the free carboxylic headgroup is necessary. The inhibition potency did not correlate with the fatty acid effects on the bilayer deformation energies. AA does not affect the conformational dynamics of the protein, suggesting it does not inhibit structural transitions necessary for transport. Single-transporter and membrane voltage assays showed that AA and related fatty acids mediate cation leak, dissipating the driving sodium gradient. Thus, such fatty acids can act as cation ionophores, suggesting a general modulatory mechanism of membrane channels and ion-coupled transporters.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Department of Physiology & Biophysics, Weill Cornell Medicine, New York, New York, USA.
| | - Radda Rusinova
- Department of Physiology & Biophysics, Weill Cornell Medicine, New York, New York, USA
| | - G Glenn Gregorio
- Department of Physiology & Biophysics, Weill Cornell Medicine, New York, New York, USA; Howard Hughes Medical Institute, Weill Cornell Medicine, New York, New York, USA
| | - Olga Boudker
- Department of Physiology & Biophysics, Weill Cornell Medicine, New York, New York, USA; Howard Hughes Medical Institute, Weill Cornell Medicine, New York, New York, USA.
| |
Collapse
|
18
|
Salis Torres A, Lee JE, Caporali A, Semple RK, Horrocks MH, MacRae VE. Mitochondrial Dysfunction as a Potential Mechanism Mediating Cardiac Comorbidities in Parkinson's Disease. Int J Mol Sci 2024; 25:10973. [PMID: 39456761 PMCID: PMC11507255 DOI: 10.3390/ijms252010973] [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: 09/11/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Individuals diagnosed with Parkinson's disease (PD) often exhibit heightened susceptibility to cardiac dysfunction, reflecting a complex interaction between these conditions. The involvement of mitochondrial dysfunction in the development and progression of cardiac dysfunction and PD suggests a plausible commonality in some aspects of their molecular pathogenesis, potentially contributing to the prevalence of cardiac issues in PD. Mitochondria, crucial organelles responsible for energy production and cellular regulation, play important roles in tissues with high energetic demands, such as neurons and cardiac cells. Mitochondrial dysfunction can occur in different and non-mutually exclusive ways; however, some mechanisms include alterations in mitochondrial dynamics, compromised bioenergetics, biogenesis deficits, oxidative stress, impaired mitophagy, and disrupted calcium balance. It is plausible that these factors contribute to the increased prevalence of cardiac dysfunction in PD, suggesting mitochondrial health as a potential target for therapeutic intervention. This review provides an overview of the physiological mechanisms underlying mitochondrial quality control systems. It summarises the diverse roles of mitochondria in brain and heart function, highlighting shared pathways potentially exhibiting dysfunction and driving cardiac comorbidities in PD. By highlighting strategies to mitigate dysfunction associated with mitochondrial impairment in cardiac and neural tissues, our review aims to provide new perspectives on therapeutic approaches.
Collapse
Affiliation(s)
- Agustina Salis Torres
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RH, UK; (A.S.T.); (J.-E.L.)
- EaStCHEM School of Chemistry, University of Edinburgh, Edinburgh EH9 3FJ, UK;
| | - Ji-Eun Lee
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RH, UK; (A.S.T.); (J.-E.L.)
- IRR Chemistry Hub, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Andrea Caporali
- Centre for Cardiovascular Science, Queen’s Medical Research Institute (QMRI), The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK; (A.C.); (R.K.S.)
| | - Robert K. Semple
- Centre for Cardiovascular Science, Queen’s Medical Research Institute (QMRI), The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK; (A.C.); (R.K.S.)
| | - Mathew H. Horrocks
- EaStCHEM School of Chemistry, University of Edinburgh, Edinburgh EH9 3FJ, UK;
- MRC Human Genetics Unit, Institute for Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Vicky E. MacRae
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RH, UK; (A.S.T.); (J.-E.L.)
| |
Collapse
|
19
|
Liang S, Zhao Z, Liu L, Zhang Y, Liu X. Research Progress on the Mechanisms of Protocatechuic Acid in the Treatment of Cognitive Impairment. Molecules 2024; 29:4724. [PMID: 39407652 PMCID: PMC11478363 DOI: 10.3390/molecules29194724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/25/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
Cognitive impairment (CI) is a type of mental health disorder that mainly affects cognitive abilities, such as learning, memory, perception, and problem-solving. Currently, in clinical practice, the treatment of cognitive impairment mainly focuses on the application of cholinesterase inhibitors and NMDA receptor antagonists; however, there is no specific and effective drug yet. Procatechuic acid (PCA) possesses various functions, including antibacterial, antiasthmatic, and expectorant effects. In recent years, it has received growing attention in the cognitive domain. Therefore, by summarizing the mechanisms of action of procatechuic acid in the treatment of cognitive impairment in this paper, it is found that procatechuic acid has multiple effects, such as regulating the expression of neuroprotective factors, inhibiting cell apoptosis, promoting the autophagy-lysosome pathway, suppressing oxidative stress damage, inhibiting inflammatory responses, improving synaptic plasticity dysfunction, inhibiting Aβ deposition, reducing APP hydrolysis, enhancing the cholinergic system, and inhibiting the excitotoxicity of neuronal cells. The involved signaling pathways include activating Pi3K-akt-mTor and inhibiting JNK, P38 MAPK, P38-ERK-JNK, SIRT1, and NF-κB/p53, etc. This paper aims to present the latest progress in research on procatechuic acid, including aspects such as its chemical properties, sources, pharmacokinetics, mechanisms for treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Shuzhi Liang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250300, China; (S.L.)
| | - Zhongmin Zhao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250300, China; (S.L.)
| | - Leilei Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250300, China; (S.L.)
| | - Yan Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250300, China; (S.L.)
- The Youth Research and Innovation Team of TCM for the Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Shandong University of Traditional Chinese Medicine, Jinan 250300, China
| | - Xijian Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250300, China; (S.L.)
| |
Collapse
|
20
|
Kasper J, Caspers S, Lotter LD, Hoffstaedter F, Eickhoff SB, Dukart J. Resting-State Changes in Aging and Parkinson's Disease Are Shaped by Underlying Neurotransmission: A Normative Modeling Study. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2024; 9:986-997. [PMID: 38679325 DOI: 10.1016/j.bpsc.2024.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/15/2024] [Accepted: 04/16/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Human healthy and pathological aging is linked to a steady decline in brain resting-state activity and connectivity measures. The neurophysiological mechanisms that underlie these changes remain poorly understood. METHODS Making use of recent developments in normative modeling and availability of in vivo maps for various neurochemical systems, we tested in the UK Biobank cohort (n = 25,917) whether and how age- and Parkinson's disease-related resting-state changes in commonly applied local and global activity and connectivity measures colocalize with underlying neurotransmitter systems. RESULTS We found that the distributions of several major neurotransmitter systems including serotonergic, dopaminergic, noradrenergic, and glutamatergic neurotransmission correlated with age-related changes across functional activity and connectivity measures. Colocalization patterns in Parkinson's disease deviated from normative aging trajectories for these, as well as for cholinergic and GABAergic (gamma-aminobutyric acidergic) neurotransmission. The deviation from normal colocalization of brain function and GABAA correlated with disease duration. CONCLUSIONS These findings provide new insights into molecular mechanisms underlying age- and Parkinson's-related brain functional changes by extending the existing evidence elucidating the vulnerability of specific neurochemical attributes to normal aging and Parkinson's disease. The results particularly indicate that alongside dopamine and serotonin, increased vulnerability of glutamatergic, cholinergic, and GABAergic systems may also contribute to Parkinson's disease-related functional alterations. Combining normative modeling and neurotransmitter mapping may aid future research and drug development through deeper understanding of neurophysiological mechanisms that underlie specific clinical conditions.
Collapse
Affiliation(s)
- Jan Kasper
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany; Institute of Neuroscience and Medicine (INM-7), Research Centre Jülich, Jülich, Germany
| | - Svenja Caspers
- Institute for Anatomy I, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany; Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
| | - Leon D Lotter
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany; Institute of Neuroscience and Medicine (INM-7), Research Centre Jülich, Jülich, Germany; Max Planck School of Cognition, Leipzig, Germany
| | - Felix Hoffstaedter
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany; Institute of Neuroscience and Medicine (INM-7), Research Centre Jülich, Jülich, Germany
| | - Simon B Eickhoff
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany; Institute of Neuroscience and Medicine (INM-7), Research Centre Jülich, Jülich, Germany
| | - Juergen Dukart
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany; Institute of Neuroscience and Medicine (INM-7), Research Centre Jülich, Jülich, Germany.
| |
Collapse
|
21
|
Castillo-Vazquez SK, Massieu L, Rincón-Heredia R, García-de la Torre P, Quiroz-Baez R, Gomez-Verjan JC, Rivero-Segura NA. Glutamatergic Neurotransmission in Aging and Neurodegenerative Diseases: A Potential Target to Improve Cognitive Impairment in Aging. Arch Med Res 2024; 55:103039. [PMID: 38981341 DOI: 10.1016/j.arcmed.2024.103039] [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: 01/11/2024] [Revised: 06/10/2024] [Accepted: 06/26/2024] [Indexed: 07/11/2024]
Abstract
Aging is characterized by the decline in many of the individual's capabilities. It has been recognized that the brain undergoes structural and functional changes during aging that are occasionally associated with the development of neurodegenerative diseases. In this sense, altered glutamatergic neurotransmission, which involves the release, binding, reuptake, and degradation of glutamate (Glu) in the brain, has been widely studied in physiological and pathophysiological aging. In particular, changes in glutamatergic neurotransmission are exacerbated during neurodegenerative diseases and are associated with cognitive impairment, characterized by difficulties in memory, learning, concentration, and decision-making. Thus, in the present manuscript, we aim to highlight the relevance of glutamatergic neurotransmission during cognitive impairment to develop novel strategies to prevent, ameliorate, or delay cognitive decline. To achieve this goal, we provide a comprehensive review of the changes reported in glutamatergic neurotransmission components, such as Glu transporters and receptors during physiological aging and in the most studied neurodegenerative diseases. Finally, we describe the current therapeutic strategies developed to target glutamatergic neurotransmission.
Collapse
Affiliation(s)
- Selma Karime Castillo-Vazquez
- Dirección de Investigación, Instituto Nacional de Geriatría, Mexico City, Mexico; Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Lourdes Massieu
- Departamento de Neuropatología Molecular, División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Ruth Rincón-Heredia
- Unidad de Imagenología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | - Paola García-de la Torre
- 4 Unidad de Investigación Epidemiológica y en Servicios de Salud, Área de Envejecimiento, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City Mexico
| | - Ricardo Quiroz-Baez
- Dirección de Investigación, Instituto Nacional de Geriatría, Mexico City, Mexico
| | | | | |
Collapse
|
22
|
Ulger O, Eş I, Proctor CM, Algin O. Stroke studies in large animals: Prospects of mitochondrial transplantation and enhancing efficiency using hydrogels and nanoparticle-assisted delivery. Ageing Res Rev 2024; 100:102469. [PMID: 39191353 DOI: 10.1016/j.arr.2024.102469] [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: 06/12/2024] [Revised: 08/08/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024]
Abstract
One of the most frequent reasons for mortality and disability today is acute ischemic stroke, which occurs by an abrupt disruption of cerebral circulation. The intricate damage mechanism involves several factors, such as inflammatory response, disturbance of ion balance, loss of energy production, excessive reactive oxygen species and glutamate release, and finally, neuronal death. Stroke research is now carried out using several experimental models and potential therapeutics. Furthermore, studies are being conducted to address the shortcomings of clinical care. A great deal of research is being done on novel pharmacological drugs, mitochondria targeting compounds, and different approaches including brain cooling and new technologies. Still, there are many unanswered questions about disease modeling and treatment strategies. Before these new approaches may be used in therapeutic settings, they must first be tested on large animals, as most of them have been done on rodents. However, there are several limitations to large animal stroke models used for research. In this review, the damage mechanisms in acute ischemic stroke and experimental acute ischemic stroke models are addressed. The current treatment approaches and promising experimental methods such as mitochondrial transplantation, hydrogel-based interventions, and strategies like mitochondria encapsulation and chemical modification, are also examined in this work.
Collapse
Affiliation(s)
- Oner Ulger
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, Ankara 06010, Turkiye; Gulhane Training and Research Hospital, University of Health Sciences, Ankara 06010, Turkiye.
| | - Ismail Eş
- Department of Engineering Science, Institute of Biomedical Engineering (IBME), University of Oxford, Oxford OX3 7DQ, UK
| | - Christopher M Proctor
- Department of Engineering Science, Institute of Biomedical Engineering (IBME), University of Oxford, Oxford OX3 7DQ, UK
| | - Oktay Algin
- Interventional MR Clinical R&D Institute, Ankara University, Ankara 06100, Turkiye; Department of Radiology, Medical Faculty, Ankara University, Ankara 06100, Turkiye; National MR Research Center (UMRAM), Bilkent University, Ankara 06800, Turkiye
| |
Collapse
|
23
|
Jiang Y, Wu W, Huang J, Liu N, Wang J, Wan X, Qin Z, Wang Y. KA-mediated excitotoxicity induces neuronal ferroptosis through activation of ferritinophagy. CNS Neurosci Ther 2024; 30:e70054. [PMID: 39306799 PMCID: PMC11416743 DOI: 10.1111/cns.70054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 08/13/2024] [Accepted: 09/06/2024] [Indexed: 09/25/2024] Open
Abstract
OBJECTIVES This study aims to elucidate the role of Fe2+ overload in kainic acid (KA)-induced excitotoxicity, investigate the involvement of ferritinophagy selective cargo receptor NCOA4 in the pathogenesis of excitotoxicity. METHODS Western blotting was used to detect the expression of FTH1, NCOA4, Lamp2, TfR, FPN, and DMT1 after KA stereotaxic injection into the unilateral striatum of mice. Colocalization of Fe2+ with lysosomes in KA-treated primary cortical neurons was observed by using confocal microscopy. Desferrioxamine (DFO) was added to chelate free iron, a CCK8 kit was used to measure cell viability, and the Fe2+ levels were detected by FerroOrange. BODIPY C11 was used to determine intracellular lipid reactive oxygen species (ROS) levels, and the mRNA levels of PTGS2, a biomarker of ferroptosis, were measured by fluorescent quantitative PCR. 3-Methyladenine (3-MA) was employed to inhibit KA-induced activation of autophagy, and changes in ferritinophagy-related protein expression and the indicated biomarkers of ferroptosis were detected. Endogenous NCOA4 was knocked down by lentivirus transfection, and cell viability and intracellular Fe2+ levels were observed after KA treatment. RESULTS Western blot results showed that the expression of NCOA4, DMT1, and Lamp2 was significantly upregulated, while FTH1 was downregulated, but there were no significant changes in TfR and FPN. The fluorescence results indicated that KA enhanced the colocalization of free Fe2+ with lysosomes in neurons. DFO intervention could effectively rescue cell damage, reduce intracellular lipid peroxidation, and decrease the increased transcript levels of PTGS2 caused by KA. Pretreatment with 3-MA effectively reversed KA-induced ferritinophagy and ferroptosis. Endogenous interference with NCOA4 significantly improved cell viability and reduced intracellular free Fe2+ levels in KA-treated cells. CONCLUSION KA-induced excitotoxicity activates ferritinophagy, and targeting ferritinophagy effectively inhibits downstream ferroptosis. Interference with NCOA4 effectively attenuates KA-induced neuronal damage. This study provides a potential therapeutic target for excitotoxicity related disease conditions.
Collapse
Affiliation(s)
- Yi‐Yue Jiang
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric DiseasesSoochow UniversitySuzhouChina
| | - Wei‐Long Wu
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric DiseasesSoochow UniversitySuzhouChina
| | - Jia‐Ni Huang
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric DiseasesSoochow UniversitySuzhouChina
| | - Na Liu
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric DiseasesSoochow UniversitySuzhouChina
| | - Jing Wang
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric DiseasesSoochow UniversitySuzhouChina
| | - Xiao‐Rui Wan
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric DiseasesSoochow UniversitySuzhouChina
| | - Zheng‐Hong Qin
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric DiseasesSoochow UniversitySuzhouChina
| | - Yan Wang
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric DiseasesSoochow UniversitySuzhouChina
| |
Collapse
|
24
|
Moloney RA, Palliser HK, Pavy CL, Shaw JC, Hirst JJ. Zuranolone therapy protects frontal cortex neurodevelopment and improves behavioral outcomes after preterm birth. Brain Behav 2024; 14:e70009. [PMID: 39236116 PMCID: PMC11376442 DOI: 10.1002/brb3.70009] [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: 03/26/2024] [Revised: 07/30/2024] [Accepted: 08/11/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Preterm birth is associated with brain injury and long-term behavioral abnormalities, for which there are limited prevention options. When born preterm, infants prematurely lose placental neurosteroid (allopregnanolone) support. This increases the risk of excitotoxic damage to the brain, which increases the risk of injury, causing long-term deficits in behavior, myelination, and alterations to neurotransmitter pathways. We propose that postnatal restoration of neurosteroid action through zuranolone therapy will reduce neurological impairments following preterm birth. METHODS Guinea pig dams underwent survival cesarean section surgery to deliver pups prematurely (GA64) or at term (GA69). Between birth and term equivalence age, preterm pups received vehicle (15% β-cyclodextrin) or the allopregnanolone analogue zuranolone (1 mg/kg/day). Behavioral analysis was performed at postnatal day (PND) 7 and 40, before tissue collection at PND 42. Immunostaining for myelin basic protein (MBP), as well as real-time polymerase chain reaction to characterize oligodendrocyte lineage and neurotransmitter pathways, was performed in frontal cortex tissues. RESULTS Zuranolone treatment prevented the hyperactive phenotype in preterm-born offspring, most markedly in males. Additionally, preterm-related reductions in MBP were ameliorated. Several preterm-related alterations in mRNA expression of dopaminergic, glutamatergic, and GABAergic pathways were also restored back to that of a term control level. CONCLUSION This is the first study to assess zuranolone treatment as a neuroprotective therapy following preterm birth. Zuranolone treatment improved behavioral outcomes and structural changes in the preterm offspring, which continued long term until at least a late childhood timepoint. Clinical studies are warranted for further exploring the neuroprotective possibilities of this treatment following preterm birth.
Collapse
Affiliation(s)
- Roisin A Moloney
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Hunter Medical Research Institute, Mothers and Babies Research Centre, Newcastle, Australia
| | - Hannah K Palliser
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Hunter Medical Research Institute, Mothers and Babies Research Centre, Newcastle, Australia
| | - Carlton L Pavy
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Hunter Medical Research Institute, Mothers and Babies Research Centre, Newcastle, Australia
| | - Julia C Shaw
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Hunter Medical Research Institute, Mothers and Babies Research Centre, Newcastle, Australia
| | - Jonathan J Hirst
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Hunter Medical Research Institute, Mothers and Babies Research Centre, Newcastle, Australia
| |
Collapse
|
25
|
Dunot J, Moreno S, Gandin C, Pousinha PA, Amici M, Dupuis J, Anisimova M, Winschel A, Uriot M, Petshow SJ, Mensch M, Bethus I, Giudici C, Hampel H, Wefers B, Wurst W, Naumann R, Ashby MC, Laube B, Zito K, Mellor JR, Groc L, Willem M, Marie H. APP fragment controls both ionotropic and non-ionotropic signaling of NMDA receptors. Neuron 2024; 112:2708-2720.e9. [PMID: 38878768 PMCID: PMC11343662 DOI: 10.1016/j.neuron.2024.05.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 04/09/2024] [Accepted: 05/29/2024] [Indexed: 07/20/2024]
Abstract
NMDA receptors (NMDARs) are ionotropic receptors crucial for brain information processing. Yet, evidence also supports an ion-flux-independent signaling mode mediating synaptic long-term depression (LTD) and spine shrinkage. Here, we identify AETA (Aη), an amyloid-β precursor protein (APP) cleavage product, as an NMDAR modulator with the unique dual regulatory capacity to impact both signaling modes. AETA inhibits ionotropic NMDAR activity by competing with the co-agonist and induces an intracellular conformational modification of GluN1 subunits. This favors non-ionotropic NMDAR signaling leading to enhanced LTD and favors spine shrinkage. Endogenously, AETA production is increased by in vivo chemogenetically induced neuronal activity. Genetic deletion of AETA production alters NMDAR transmission and prevents LTD, phenotypes rescued by acute exogenous AETA application. This genetic deletion also impairs contextual fear memory. Our findings demonstrate AETA-dependent NMDAR activation (ADNA), characterizing AETA as a unique type of endogenous NMDAR modulator that exerts bidirectional control over NMDAR signaling and associated information processing.
Collapse
Affiliation(s)
- Jade Dunot
- Université Côte d'Azur, CNRS, INSERM, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France; Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Sebastien Moreno
- Université Côte d'Azur, CNRS, INSERM, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Carine Gandin
- Université Côte d'Azur, CNRS, INSERM, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Paula A Pousinha
- Université Côte d'Azur, CNRS, INSERM, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Mascia Amici
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| | - Julien Dupuis
- Université de Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, 33076 Bordeaux Cedex, France
| | - Margarita Anisimova
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
| | - Alex Winschel
- Department of Biology, Neurophysiology und Neurosensory Systems, TU Darmstadt, 64287 Darmstadt, Germany
| | - Magalie Uriot
- Université de Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, 33076 Bordeaux Cedex, France
| | - Samuel J Petshow
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
| | - Maria Mensch
- Université Côte d'Azur, CNRS, INSERM, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Ingrid Bethus
- Université Côte d'Azur, CNRS, INSERM, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Camilla Giudici
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Heike Hampel
- Biomedical Center (BMC), Division of Metabolic Biochemistry, Faculty of Medicine, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Benedikt Wefers
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany; Institute of Developmental Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Wolfgang Wurst
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany; Institute of Developmental Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Ronald Naumann
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Michael C Ashby
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| | - Bodo Laube
- Department of Biology, Neurophysiology und Neurosensory Systems, TU Darmstadt, 64287 Darmstadt, Germany
| | - Karen Zito
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
| | - Jack R Mellor
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| | - Laurent Groc
- Université de Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, 33076 Bordeaux Cedex, France
| | - Michael Willem
- Biomedical Center (BMC), Division of Metabolic Biochemistry, Faculty of Medicine, Ludwig-Maximilians-Universität München, 81377 Munich, Germany.
| | - Hélène Marie
- Université Côte d'Azur, CNRS, INSERM, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France.
| |
Collapse
|
26
|
Dhureja M, Deshmukh R. Impact of alogliptin on lipopolysaccharide-induced experimental Parkinson's disease: Unrevealing neurochemical and histopathological alterations in rodents. Eur J Pharmacol 2024; 975:176635. [PMID: 38734296 DOI: 10.1016/j.ejphar.2024.176635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/04/2024] [Accepted: 05/03/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Degeneration of the nigrostriatal dopaminergic pathway has been seen as a significant cause of movement disability in Parkinson's disease (PD) patients. However, the exact reason for these degenerative changes has remained obscure. In recent years, incretins have been neuroprotective in various pathologies. In the current study, we have investigated the neuroprotective potential of alogliptin (Alo), a dipeptidyl peptidase-IV (DPP-IV) inhibitor, in a lipopolysaccharide (LPS) induced experimental model of PD. EXPERIMENTAL APPROACH LPS (5μg/5 μl) was infused intranigrally to induce PD in experimental rats. Post-LPS infusion, these animals were treated with Alo for 21 days in three successive dosages of 10, 20, and 40 mg/kg/day/per oral. The study is well supported with the determinations of motor functions biochemical, neurochemical, and histological analysis. KEY RESULTS Intranigral infusion of LPS in rats produced motor deficit. It was accompanied by oxidative stress, elevation in neuroinflammatory cytokines, altered neurochemistry, and degenerative changes in the striatal brain region. While Alo abrogated LPS-induced biochemical/neurochemical alterations, improved motor functions, and preserved neuronal morphology in LPS-infused rats. CONCLUSION The observed neuroprotective potential of Alo may be due to its antioxidant and anti-inflammatory actions and its ability to modulate monoaminergic signals. Nonetheless, current findings suggest that improving the availability of incretins through DPP-IV inhibition is a promising strategy for treating Parkinson's disease.
Collapse
Affiliation(s)
- Maanvi Dhureja
- Department of Pharmaceutical Sciences & Technology, MRSPTU, Bathinda, India
| | - Rahul Deshmukh
- Department of Pharmaceutical Sciences & Technology, MRSPTU, Bathinda, India; Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India.
| |
Collapse
|
27
|
Marvi PK, Ahmed SR, Das P, Ghosh R, Srinivasan S, Rajabzadeh AR. Prunella vulgaris-phytosynthesized platinum nanoparticles: Insights into nanozymatic activity for H 2O 2 and glutamate detection and antioxidant capacity. Talanta 2024; 274:125998. [PMID: 38574541 DOI: 10.1016/j.talanta.2024.125998] [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: 11/14/2023] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/06/2024]
Abstract
Artificial nanozymes (enzyme-mimics), specifically metallic nanomaterials, have garnered significant attention recently due to their reduced preparation cost and enhanced stability in a wide range of environments. The present investigation highlights, for the first time, a straightforward green synthesis of biogenic platinum nanoparticles (PtNPs) from a natural resource, namely Prunella vulgaris (Pr). To demonstrate the effectiveness of the phytochemical extract as an effective reducing agent, the PtNPs were characterized by various techniques such as UV-vis spectroscopy, High-resolution Transmission electron microscopy (HR-TEM), zeta-potential analysis, Fourier-transform infrared spectroscopy (FTIR), and Energy dispersive spectroscopy (EDS). The formation of PtNPs with narrow size distribution was verified. Surface decoration of PtNPs was demonstrated with multitudinous functional groups springing from the herbal extract. To demonstrate their use as viable nanozymes, the peroxidase-like activity of Pr/PtNPs was evaluated through a colorimetric assay. Highly sensitive visual detection of H2O2 with discrete linear ranges and a low detection limit of 3.43 μM was demonstrated. Additionally, peroxidase-like catalytic activity was leveraged to develop a colorimetric platform to quantify glutamate biomarker levels with a high degree of selectivity, the limit of detection (LOD) being 7.00 μM. The 2,2-Diphenyl-1-picrylhydrazyl (DPPH) test was used to explore the scavenging nature of the PtNPs via the degradation of DPPH. Overall, the colorimetric assay developed using the Pr/PtNP nanozymes in this work could be used in a broad spectrum of applications, ranging from biomedicine and food science to environmental monitoring.
Collapse
Affiliation(s)
- Parham Khoshbakht Marvi
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L7, Canada
| | - Syed Rahin Ahmed
- School of Engineering Practice and Technology, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L7, Canada
| | - Poushali Das
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L7, Canada
| | - Raja Ghosh
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L7, Canada; Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L7, Canada
| | - Seshasai Srinivasan
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L7, Canada; School of Engineering Practice and Technology, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L7, Canada.
| | - Amin Reza Rajabzadeh
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L7, Canada; School of Engineering Practice and Technology, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L7, Canada.
| |
Collapse
|
28
|
Low ZY, Yip AJW, Chan AML, Choo WS. 14-3-3 Family of Proteins: Biological Implications, Molecular Interactions, and Potential Intervention in Cancer, Virus and Neurodegeneration Disorders. J Cell Biochem 2024; 125:e30624. [PMID: 38946063 DOI: 10.1002/jcb.30624] [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: 03/01/2024] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 07/02/2024]
Abstract
The 14-3-3 family of proteins are highly conserved acidic eukaryotic proteins (25-32 kDa) abundantly present in the body. Through numerous binding partners, the 14-3-3 is responsible for many essential cellular pathways, such as cell cycle regulation and gene transcription control. Hence, its dysregulation has been linked to the onset of critical illnesses such as cancers, neurodegenerative diseases and viral infections. Interestingly, explorative studies have revealed an inverse correlation of 14-3-3 protein in cancer and neurodegenerative diseases, and the direct manipulation of 14-3-3 by virus to enhance infection capacity has dramatically extended its significance. Of these, COVID-19 has been linked to the 14-3-3 proteins by the interference of the SARS-CoV-2 nucleocapsid (N) protein during virion assembly. Given its predisposition towards multiple essential host signalling pathways, it is vital to understand the holistic interactions between the 14-3-3 protein to unravel its potential therapeutic unit in the future. As such, the general structure and properties of the 14-3-3 family of proteins, as well as their known biological functions and implications in cancer, neurodegeneration, and viruses, were covered in this review. Furthermore, the potential therapeutic target of 14-3-3 proteins in the associated diseases was discussed.
Collapse
Affiliation(s)
- Zheng Yao Low
- School of Science, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Ashley Jia Wen Yip
- School of Science, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Alvin Man Lung Chan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
| | - Wee Sim Choo
- School of Science, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| |
Collapse
|
29
|
Ergul Erkec O, Acikgoz E, Huyut Z, Akyol ME, Ozyurt EO, Keskin S. Ghrelin ameliorates neuronal damage, oxidative stress, inflammatory parameters, and GFAP expression in traumatic brain injury. Brain Inj 2024; 38:514-523. [PMID: 38433464 DOI: 10.1080/02699052.2024.2324012] [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: 05/27/2023] [Accepted: 02/22/2024] [Indexed: 03/05/2024]
Abstract
OBJECTIVE This study investigated the effects of ghrelin on oxidative stress, working memory, inflammatory parameters, and neuron degeneration. METHODS TBI was produced with the weight-drop technique. Rats in the G+TBI and TBI+G groups received ghrelin for 7 or 2 days, respectively. The control group received saline. On the 8th day of the study, the brain and blood tissue were taken under anesthesia. RESULTS A significant increase in brain GSH-PX, MDA, IL-1β, TGF-β1, and IL-8 levels and a significant decrease in CAT levels were found in the TBI group compared to the control. Serum MDA, GSH, IL-1β, and IL-8 levels were increased with TBI. Ghrelin treatment after TBI significantly increased the serum GSH, CAT, GSH-PX, and brain GSH and CAT levels, while it significantly decreased the serum MDA, IL-1β, and brain MDA, TGF-β1, and IL-8 levels. Histological evaluations revealed that ghrelin treatment led to a reduction in inflammation, while also significantly ameliorating TBI-induced neuron damage and vascular injuries. Immunohistochemistry staining showed that GFAP staining intensity was significantly increased in the cortex and hippocampus in TBI, and GFAP immunoreactivity was decreased with ghrelin treatment. CONCLUSION The results from this study suggested that ghrelin may have curative effects on TBI.
Collapse
Affiliation(s)
- Ozlem Ergul Erkec
- Department of Physiology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Eda Acikgoz
- Department of Histology and Embryology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Zubeyir Huyut
- Department of Biochemistry, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Mehmet Edip Akyol
- Department of Neurosurgery, Faculty of Mecine, Van Yuzuncu Yil University, Van, Turkey
| | | | - Sıddık Keskin
- Department of Biostatistics, Van Yuzuncu Yıl University, Van, Turkey
| |
Collapse
|
30
|
Jaiswal A, Patel M, Naseer A, Kumari S, Revi N, Rengan A, Jain A, Nazir A, Gour N, Verma S. Amyloid Mimicking Assemblies Formed by Glutamine, Glutamic Acid, and Aspartic Acid. ACS Chem Neurosci 2024; 15:2253-2264. [PMID: 38768265 DOI: 10.1021/acschemneuro.4c00082] [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: 05/22/2024] Open
Abstract
The aggregation of amino acids into amyloid-like structures is a critical phenomenon for understanding the pathophysiology of various diseases, including inborn errors of metabolism (IEMs) associated with amino acid imbalances. Previous studies have primarily focused on self-assembly of aromatic amino acids, leading to a limited understanding of nonaromatic, polar amino acids in this context. To bridge this gap, our study investigates the self-assembly and aggregation behavior of specific nonaromatic charged and uncharged polar amino acids l-glutamine (Gln), l-aspartic acid (Asp), and l-glutamic acid (Glu), which have not been reported widely in the context of amyloid aggregation. Upon aging these amino acids under controlled conditions, we observed the formation of uniform, distinct aggregates, with Gln forming fibrillar gel-like structures and Glu exhibiting fibrous globular morphologies. Computational simulations validated these findings, identifying Gln as the most potent in forming stable aggregates, followed by Glu and Asp. These simulations elucidated the driving forces behind the distinct morphologies and stabilities of the aggregates. Thioflavin T assays were employed to confirm the amyloid-like nature of these aggregates, suggesting their potential cytotoxic impact. To assess toxicity, we performed in vitro studies on neural cell lines and in vivo experiments in Caenorhabditis elegans (C. elegans), which demonstrated measurable cytotoxic effects, corroborated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide and heat shock survival assays. Importantly, this study fills a critical gap in our understanding on the role of nonaromatic amino acids in amyloidogenesis and its implications for IEMs. Our findings provide a foundation for future investigations into the mechanisms of diseases associated with amino acid accumulation and offer potential avenues for the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Ankita Jaiswal
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - Monisha Patel
- School of Science, Indrashil University, Kadi, Mehsana, Gujarat 382740, India
| | - Anam Naseer
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Division of Toxicology & Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Simran Kumari
- Department of Bioengineering and Biotechnology, Birla Institute of Technology Mesra, Ranchi 835215, Jharkhand, India
| | - Neeraja Revi
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Hyderabad, Telangana 502285, India
| | - Aravind Rengan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Hyderabad, Telangana 502285, India
| | - Alok Jain
- Department of Bioengineering and Biotechnology, Birla Institute of Technology Mesra, Ranchi 835215, Jharkhand, India
| | - Aamir Nazir
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Division of Toxicology & Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Nidhi Gour
- School of Science, Indrashil University, Kadi, Mehsana, Gujarat 382740, India
| | - Sandeep Verma
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| |
Collapse
|
31
|
Doppler CEJ, Seger A, Farrher E, Régio Brambilla C, Hensel L, Filss CP, Hellmich M, Gogishvili A, Shah NJ, Lerche CW, Neumaier B, Langen KJ, Fink GR, Sommerauer M. Glutamate Signaling in Patients With Parkinson Disease With REM Sleep Behavior Disorder. Neurology 2024; 102:e209271. [PMID: 38630966 DOI: 10.1212/wnl.0000000000209271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Clinical heterogeneity of patients with Parkinson disease (PD) is well recognized. PD with REM sleep behavior disorder (RBD) is a more malignant phenotype with faster motor progression and higher nonmotor symptom burden. However, the neural mechanisms underlying this clinical divergence concerning imbalances in neurotransmitter systems remain elusive. METHODS Combining magnetic resonance (MR) spectroscopy and [11C]ABP688 PET on a PET/MR hybrid system, we simultaneously investigated two different mechanisms of glutamate signaling in patients with PD. Patients were grouped according to their RBD status in overnight video-polysomnography and compared with age-matched and sex-matched healthy control (HC) participants. Total volumes of distribution (VT) of [11C]ABP688 were estimated with metabolite-corrected plasma concentrations during steady-state conditions between 45 and 60 minutes of the scan following a bolus-infusion protocol. Glutamate, glutamine, and glutathione levels were investigated with single-voxel stimulated echo acquisition mode MR spectroscopy of the left basal ganglia. RESULTS We measured globally elevated VT of [11C]ABP688 in 16 patients with PD and RBD compared with 17 patients without RBD and 15 HC participants (F(2,45) = 5.579, p = 0.007). Conversely, glutamatergic metabolites did not differ between groups and did not correlate with the regional VT of [11C]ABP688. VT of [11C]ABP688 correlated with the amount of REM sleep without atonia (F(1,42) = 5.600, p = 0.023) and with dopaminergic treatment response in patients with PD (F(1,30) = 5.823, p = 0.022). DISCUSSION Our results suggest that patients with PD and RBD exhibit altered glutamatergic signaling indicated by higher VT of [11C]ABP688 despite unaffected glutamate levels. The imbalance of glutamate receptors and MR spectroscopy glutamate metabolite levels indicates a novel mechanism contributing to the heterogeneity of PD and warrants further investigation of drugs targeting mGluR5.
Collapse
Affiliation(s)
- Christopher E J Doppler
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Aline Seger
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Ezequiel Farrher
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Cláudia Régio Brambilla
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Lukas Hensel
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Christian P Filss
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Martin Hellmich
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Ana Gogishvili
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - N Jon Shah
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Christoph W Lerche
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Bernd Neumaier
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Karl-Josef Langen
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Gereon R Fink
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Michael Sommerauer
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| |
Collapse
|
32
|
Liu L, Tian X, Li W. Mechanistic study of the anti-excitatory amino acid toxicity of Bushen Zhichan decoction for Parkinson's disease based on the transcriptional regulation of EAAT1 by YY1. JOURNAL OF ETHNOPHARMACOLOGY 2024; 325:117857. [PMID: 38350506 DOI: 10.1016/j.jep.2024.117857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/25/2024] [Accepted: 02/02/2024] [Indexed: 02/15/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bushen Zhichan decoction (BSZCF) is derived from Liuwei Dihuang Pill, a famous Chinese herbal formula recorded in the book Key to Therapeutics of Children's Diseases. It has been widely used as a basic prescription for nourishing and tonifying the liver and kidneys to treat Parkinson's disease (PD), but its mechanism remains to be explored. AIM OF THE STUDY BSZCF, a Chinese herbal formula comprising five herbs: Rehmannia glutinosa (Gaertn.) DC., Dioscorea oppositifolia L., Cornus officinalis Siebold & Zucc., Fallopia multiflora (Thunb.) Haraldson and Cistanche tubulosa (Schenk) Wight, is used clinically to treat PD. In vivo and in vitro experiments were designed to elucidate the mechanism of BSZCF in the protection of dopamine (DA) neurons and the treatment of PD. The toxicity of excitatory amino acids (EAA) may be attenuated by inhibiting the transcription factor Yin Yang 1 (YY1) and up-regulating the expression of excitatory amino acid transporter 1 (EAAT1). MATERIALS AND METHODS IN VIVO: After 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) was intraperitoneally injected into specific pathogen free (SPF) C57BL/6J mice, model mice were intragastrically given adamantane hydrochloride tablets (AHT) or different doses of BSZCF for 14 days. Both open field and pole-climbing tests were conducted to assess behavioral changes. In vitro: 1-Methyl-4-phe-nylpyridiniumiodide (MPP+)-injured human neuroblastoma cells (SH-SY5Y) were utilized to construct PD cell models. Primary astrocytes were transfected with EAAT1 and YY1 lentiviruses for EAAT1 gene knockout and YY1 gene knockout astrocytes, respectively. The high performance liquid chromatography-mass spectrometry (HPLC-MS) analysis of BSZCF was performed to control the quality of blood drugs. The optimal concentration and time of PD cell models treated by BSZCF were determined by the use of Cell Counting Kit-8 (CCK8). Enzyme-linked immunosorbent assay (ELISA) was used for measuring glutamate (Glu) in the peripheral blood and cells of each group. Western blotting (WB) and real-time quantitative polymerase chain reaction (qPCR) were used to detect tyrosine hydroxylase (TH), dopamine transporters (DAT), EAAT1 and YY1 protein and mRNA. After the blockade of EAAT1, immunofluorescence (IF) assay was used to detect the TH protein in each group. RESULTS In vivo research showed that BSZCF improved the behavioral symptoms of PD mice, and reduced the death of DA neurons and the level of Glu. The mechanism may be related to the decrease of YY1 expression and the increase of EAAT1 levels. In vitro experiments showed that the anti-excitatory amino acid toxicity of BSZCF was achieved by inhibiting YY1 expression and regulating EAAT1. CONCLUSIONS By inhibiting YY1 to increase the expression of EAAT1 and attenuating the toxicity of Glu, BSZCF exerts the effect of protecting DA neurons and treating PD-like symptoms in mice.
Collapse
Affiliation(s)
- Leilei Liu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, China.
| | - Xinyun Tian
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, China.
| | - Wentao Li
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, China.
| |
Collapse
|
33
|
Tang Y, Wu X, Li J, Li Y, Xu X, Li G, Zhang P, Qin C, Wu LJ, Tang Z, Tian DS. The Emerging Role of Microglial Hv1 as a Target for Immunomodulation in Myelin Repair. Aging Dis 2024; 15:1176-1203. [PMID: 38029392 PMCID: PMC11081154 DOI: 10.14336/ad.2023.1107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023] Open
Abstract
In the central nervous system (CNS), the myelin sheath ensures efficient interconnection between neurons and contributes to the regulation of the proper function of neuronal networks. The maintenance of myelin and the well-organized subtle process of myelin plasticity requires cooperation among myelin-forming cells, glial cells, and neural networks. The process of cooperation is fragile, and the balance is highly susceptible to disruption by microenvironment influences. Reactive microglia play a critical and complicated role in the demyelination and remyelination process. Recent studies have shown that the voltage-gated proton channel Hv1 is selectively expressed in microglia in CNS, which regulates intracellular pH and is involved in the production of reactive oxygen species, underlying multifaceted roles in maintaining microglia function. This paper begins by examining the molecular mechanisms of demyelination and emphasizes the crucial role of the microenvironment in demyelination. It focuses specifically on the role of Hv1 in myelin repair and its therapeutic potential in CNS demyelinating diseases.
Collapse
Affiliation(s)
- Yingxin Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xuan Wu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jiarui Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yuanwei Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiaoxiao Xu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Gaigai Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Ping Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
34
|
Spitz S, Schobesberger S, Brandauer K, Ertl P. Sensor-integrated brain-on-a-chip platforms: Improving the predictive validity in neurodegenerative research. Bioeng Transl Med 2024; 9:e10604. [PMID: 38818126 PMCID: PMC11135156 DOI: 10.1002/btm2.10604] [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: 07/05/2023] [Revised: 08/31/2023] [Accepted: 09/12/2023] [Indexed: 06/01/2024] Open
Abstract
Affecting millions of individuals worldwide, neurodegenerative diseases (NDDs) pose a significant and growing health concern in people over the age of 60 years. Contributing to this trend are the steady increase in the aging population coupled with a persistent lack of disease-altering treatment strategies targeting NDDs. The absence of efficient therapeutics can be attributed to high failure rates in clinical trials and the ineptness of animal models in preceding preclinical studies. To that end, in recent years, significant research effort has been dedicated to the development of human cell-based preclinical disease models characterized by a higher degree of predictive validity. However, a key requirement of any in vitro model constitutes the precise knowledge and replication of the target tissues' (patho-)physiological microenvironment. Herein, microphysiological systems have demonstrated superiority over conventional static 2D/3D in vitro cell culture systems, as they allow for the emulation and continuous monitoring of the onset, progression, and remission of disease-associated phenotypes. This review provides an overview of recent advances in the field of NDD research using organ-on-a-chip platforms. Specific focus is directed toward non-invasive sensing strategies encompassing electrical, electrochemical, and optical sensors. Additionally, promising on- and integrable off-chip sensing strategies targeting key analytes in NDDs will be presented and discussed in detail.
Collapse
Affiliation(s)
- Sarah Spitz
- Faculty of Technical ChemistryVienna University of TechnologyViennaAustria
- Present address:
Department of Mechanical Engineering and Biological EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | | | | | - Peter Ertl
- Faculty of Technical ChemistryVienna University of TechnologyViennaAustria
| |
Collapse
|
35
|
González-Cota AL, Martínez-Flores D, Rosendo-Pineda MJ, Vaca L. NMDA receptor-mediated Ca 2+ signaling: Impact on cell cycle regulation and the development of neurodegenerative diseases and cancer. Cell Calcium 2024; 119:102856. [PMID: 38408411 DOI: 10.1016/j.ceca.2024.102856] [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: 09/11/2023] [Revised: 01/08/2024] [Accepted: 02/07/2024] [Indexed: 02/28/2024]
Abstract
NMDA receptors are Ca2+-permeable ligand-gated ion channels that mediate fast excitatory transmission in the central nervous system. NMDA receptors regulate the proliferation and differentiation of neural progenitor cells and also play critical roles in neural plasticity, memory, and learning. In addition to their physiological role, NMDA receptors are also involved in glutamate-mediated excitotoxicity, which results from excessive glutamate stimulation, leading to Ca2+ overload, and ultimately to neuronal death. Thus, NMDA receptor-mediated excitotoxicity has been linked to several neurodegenerative diseases such as Alzheimer's, Parkinson's, Huntington's, dementia, and stroke. Interestingly, in addition to its effects on cell death, aberrant expression or activation of NMDA receptors is also involved in pathological cellular proliferation, and is implicated in the invasion and proliferation of various types of cancer. These disorders are thought to be related to the contribution of NMDA receptors to cell proliferation and cell death through cell cycle modulation. This review aims to discuss the evidence implicating NMDA receptor activity in cell cycle regulation and the link between aberrant NMDA receptor activity and the development of neurodegenerative diseases and cancer due to cell cycle dysregulation. The information presented here will provide insights into the signaling pathways and the contribution of NMDA receptors to these diseases, and suggests that NMDA receptors are promising targets for the prevention and treatment of these diseases, which are leading causes of death and disability worldwide.
Collapse
Affiliation(s)
- Ana L González-Cota
- Instituto de Fisiología Celular, Departamento de Biología Celular y Desarrollo, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, Ciudad de México, 04510, Mexico
| | - Daniel Martínez-Flores
- Instituto de Fisiología Celular, Departamento de Biología Celular y Desarrollo, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, Ciudad de México, 04510, Mexico
| | - Margarita Jacaranda Rosendo-Pineda
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, Ciudad de México, 04510, Mexico
| | - Luis Vaca
- Instituto de Fisiología Celular, Departamento de Biología Celular y Desarrollo, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, Ciudad de México, 04510, Mexico.
| |
Collapse
|
36
|
Matus V, Castro-Guarda M, Cárcamo-Fierro J, Morera FJ, Zambrano A. Interleukin 3 Inhibits Glutamate-Cytotoxicity in Neuroblastoma Cell Line. Neurochem Res 2024; 49:1373-1386. [PMID: 38512424 DOI: 10.1007/s11064-024-04123-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 01/06/2024] [Accepted: 02/06/2024] [Indexed: 03/23/2024]
Abstract
Interleukin 3 (IL-3) is a well-known pleiotropic cytokine that regulates the proliferation and differentiation of hematopoietic progenitor cells, triggering classical signaling pathways such as JAK/STAT, Ras/MAPK, and PI3K/Akt to carry out its functions. Interestingly, the IL-3 receptor is also expressed in non-hematopoietic cells, playing a crucial role in cell survival. Our previous research demonstrated the expression of the IL-3 receptor in neuron cells and its protective role in neurodegeneration. Glutamate, a principal neurotransmitter in the central nervous system, can induce cellular stress and lead to neurotoxicity when its extracellular concentrations surpass normal levels. This excessive glutamate presence is frequently observed in various neurological diseases. In this study, we uncover the protective role of IL-3 as an inhibitor of glutamate-induced cell death, analyzing the cytokine's signaling pathways during its protective effect. Specifically, we examined the relevance of JAK/STAT, Ras/MAPK, and PI3 K signaling pathways in the molecular mechanism triggered by IL-3. Our results show that the inhibition of JAK, ERK, and PI3 K signaling pathways, using pharmacological inhibitors, effectively blocked IL-3's protective role against glutamate-induced cell death. Additionally, our findings suggest that Bcl-2 and Bax proteins may be involved in the molecular mechanism triggered by IL-3. Our investigation into IL-3's ability to protect neuronal cells from glutamate-induced damage offers a promising therapeutic avenue with potential clinical implications for several neurological diseases characterized by glutamate neurotoxicity.
Collapse
Affiliation(s)
- Verónica Matus
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, (P. O. Box) 567, 5090000, Casilla, Valdivia, Chile
| | - Marcos Castro-Guarda
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, (P. O. Box) 567, 5090000, Casilla, Valdivia, Chile
| | - Joaquín Cárcamo-Fierro
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, (P. O. Box) 567, 5090000, Casilla, Valdivia, Chile
| | - Francisco J Morera
- Applied Biochemistry Laboratory, Escuela de Medicina Veterinaria, Facultad de Agronomía y Sistemas Naturales, Facultad de Ciencias Biológicas y Facultad de Medicina, Pontificia Universidad Católica de Chile, 7820436, Santiago, Chile
| | - Angara Zambrano
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, (P. O. Box) 567, 5090000, Casilla, Valdivia, Chile.
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile.
| |
Collapse
|
37
|
Vaglio-Garro A, Halasz A, Nováková E, Gasser AS, Zavadskis S, Weidinger A, Kozlov AV. Interplay between Energy Supply and Glutamate Toxicity in the Primary Cortical Culture. Biomolecules 2024; 14:543. [PMID: 38785950 PMCID: PMC11118065 DOI: 10.3390/biom14050543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
Limited substrate availability because of the blood-brain barrier (BBB) has made the brain develop specific molecular mechanisms to survive, using lactate synthesized by astrocytes as a source of energy in neurons. To understand if lactate improves cellular viability and susceptibility to glutamate toxicity, primary cortical cells were incubated in glucose- or lactate-containing media and toxic concentrations of glutamate for 24 h. Cell death was determined by immunostaining and lactate dehydrogenase (LDH) release. Mitochondrial membrane potential and nitric oxide (NO) levels were measured using Tetramethylrhodamine, methyl ester (TMRM) and 4-Amino-5-Methylamino-2',7'-Difluorofluorescein Diacetate (DAF-FM) live staining, respectively. LDH activity was quantified in single cells in the presence of lactate (LDH substrate) and oxamate (LDH inhibitor). Nuclei of cells were stained with DAPI and neurons with MAP2. Based on the distance between neurons and glial cells, they were classified as linked (<10 µm) and non-linked (>10 µm) neurons. Lactate increased cell death rate and the mean value of endogenous NO levels compared to glucose incubations. Mitochondrial membrane potential was lower in the cells cultured with lactate, but this effect was reversed when glutamate was added to the lactate medium. LDH activity was higher in linked neurons compared to non-linked neurons, supporting the hypothesis of the existence of the lactate shuttle between astrocytes and at least a portion of neurons. In conclusion, glucose or lactate can equally preserve primary cortical neurons, but those neurons having a low level of LDH activity and incubated with lactate cannot cover high energetic demand solely with lactate and become more susceptible to glutamate toxicity.
Collapse
Affiliation(s)
- Annette Vaglio-Garro
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria; (A.V.-G.); (A.H.); (E.N.); (A.S.G.); (S.Z.); (A.W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Andrea Halasz
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria; (A.V.-G.); (A.H.); (E.N.); (A.S.G.); (S.Z.); (A.W.)
| | - Ema Nováková
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria; (A.V.-G.); (A.H.); (E.N.); (A.S.G.); (S.Z.); (A.W.)
| | - Andreas Sebastian Gasser
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria; (A.V.-G.); (A.H.); (E.N.); (A.S.G.); (S.Z.); (A.W.)
| | - Sergejs Zavadskis
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria; (A.V.-G.); (A.H.); (E.N.); (A.S.G.); (S.Z.); (A.W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Adelheid Weidinger
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria; (A.V.-G.); (A.H.); (E.N.); (A.S.G.); (S.Z.); (A.W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Andrey V. Kozlov
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria; (A.V.-G.); (A.H.); (E.N.); (A.S.G.); (S.Z.); (A.W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
38
|
Casey C, Fullard JF, Sleator RD. Unravelling the genetic basis of Schizophrenia. Gene 2024; 902:148198. [PMID: 38266791 DOI: 10.1016/j.gene.2024.148198] [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: 09/01/2023] [Revised: 12/07/2023] [Accepted: 01/19/2024] [Indexed: 01/26/2024]
Abstract
Neuronal development is a highly regulated mechanism that is central to organismal function in animals. In humans, disruptions to this process can lead to a range of neurodevelopmental phenotypes, including Schizophrenia (SCZ). SCZ has a significant genetic component, whereby an individual with an SCZ affected family member is eight times more likely to develop the disease than someone with no family history of SCZ. By examining a combination of genomic, transcriptomic and epigenomic datasets, large-scale 'omics' studies aim to delineate the relationship between genetic variation and abnormal cellular activity in the SCZ brain. Herein, we provide a brief overview of some of the key omics methods currently being used in SCZ research, including RNA-seq, the assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) and high-throughput chromosome conformation capture (3C) approaches (e.g., Hi-C), as well as single-cell/nuclei iterations of these methods. We also discuss how these techniques are being employed to further our understanding of the genetic basis of SCZ, and to identify associated molecular pathways, biomarkers, and candidate drug targets.
Collapse
Affiliation(s)
- Clara Casey
- Department of Biological Sciences, Munster Technological University, Bishopstown, Cork, Ireland; Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - John F Fullard
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Roy D Sleator
- Department of Biological Sciences, Munster Technological University, Bishopstown, Cork, Ireland.
| |
Collapse
|
39
|
Vongthip W, Nilkhet S, Boonruang K, Sukprasansap M, Tencomnao T, Baek SJ. Neuroprotective mechanisms of luteolin in glutamate-induced oxidative stress and autophagy-mediated neuronal cell death. Sci Rep 2024; 14:7707. [PMID: 38565590 PMCID: PMC10987666 DOI: 10.1038/s41598-024-57824-2] [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: 01/24/2024] [Accepted: 03/21/2024] [Indexed: 04/04/2024] Open
Abstract
Neurodegenerative diseases, characterized by progressive neuronal dysfunction and loss, pose significant health challenges. Glutamate accumulation contributes to neuronal cell death in diseases such as Alzheimer's disease. This study investigates the neuroprotective potential of Albizia lebbeck leaf extract and its major constituent, luteolin, against glutamate-induced hippocampal neuronal cell death. Glutamate-treated HT-22 cells exhibited reduced viability, altered morphology, increased ROS, and apoptosis, which were attenuated by pre-treatment with A. lebbeck extract and luteolin. Luteolin also restored mitochondrial function, decreased mitochondrial superoxide, and preserved mitochondrial morphology. Notably, we first found that luteolin inhibited the excessive process of mitophagy via the inactivation of BNIP3L/NIX and inhibited lysosomal activity. Our study suggests that glutamate-induced autophagy-mediated cell death is attenuated by luteolin via activation of mTORC1. These findings highlight the potential of A. lebbeck as a neuroprotective agent, with luteolin inhibiting glutamate-induced neurotoxicity by regulating autophagy and mitochondrial dynamics.
Collapse
Affiliation(s)
- Wudtipong Vongthip
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Program in Clinical Biochemistry and Molecular Medicine, Chulalongkorn University, 10330, Bangkok, Thailand
- Laboratory of Signal Transduction, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Sunita Nilkhet
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Program in Clinical Biochemistry and Molecular Medicine, Chulalongkorn University, 10330, Bangkok, Thailand
- Laboratory of Signal Transduction, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Kanokkan Boonruang
- Laboratory of Signal Transduction, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Monruedee Sukprasansap
- Food Toxicology Unit, Institute of Nutrition, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Seung Joon Baek
- Laboratory of Signal Transduction, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea.
| |
Collapse
|
40
|
Roy A, Trigun SK. The restoration of hippocampal nerve de-myelination by methylcobalamin relates with the enzymatic regulation of homocysteine level in a rat model of moderate grade hepatic encephalopathy. J Biochem Mol Toxicol 2024; 38:e23695. [PMID: 38511258 DOI: 10.1002/jbt.23695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 02/08/2024] [Accepted: 03/12/2024] [Indexed: 03/22/2024]
Abstract
This article describes how methylcobalamin (MeCbl) restores nerve myelination in a moderate- grade hepatic encephalopathy (MoHE) model of ammonia neurotoxicity. The comparative profiles of myelin basic protein (MBP), homocysteine (Hcy) and methionine synthase (MS: a MeCbl- dependent enzyme) activity versus nerve myelination status were studied in the hippocampus of the control, the MoHE (developed by administering 100 mg/kg bw thioacetamide i.p. for 10 days) and the MoHE rats treated with MeCbl (500 µg/kg BW i.p.) for 7 days. Compared to those of control rats, the hippocampal CA1 and CA3 regions of the MoHE rats showed significantly lower myelinated areas and MBP immunostaining. This coincided with the deranged myelin layering in TEM images, decreased MBP protein and its transcript levels in hippocampus of MoHE rats. However, all these parameters recovered to normal levels after MeCbl treatment. MeCbl is a cofactor of MS that catalyzes the conversion of Hcy to methionine as a feeder step of methylation reactions. We observed significantly increased serum and hippocampal Hcy levels in MoHE rats, however, these levels were restored to control values with a concordant activation of MS due to MeCbl treatment. A significant recovery in neurobehavioral impairments in the MoHE rats due to MeCbl treatment was also observed. These findings suggest that MoHE pathogenesis is associated with deranged nerve myelination in the hippocampus and that MeCbl treatment is able to restore it mainly by activating MS, a MeCbl-dependent Hcy-metabolizing enzyme.
Collapse
Affiliation(s)
- Anima Roy
- Biochemistry Section, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Surendra Kumar Trigun
- Biochemistry Section, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
41
|
Amartumur S, Nguyen H, Huynh T, Kim TS, Woo RS, Oh E, Kim KK, Lee LP, Heo C. Neuropathogenesis-on-chips for neurodegenerative diseases. Nat Commun 2024; 15:2219. [PMID: 38472255 PMCID: PMC10933492 DOI: 10.1038/s41467-024-46554-8] [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: 10/04/2023] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Developing diagnostics and treatments for neurodegenerative diseases (NDs) is challenging due to multifactorial pathogenesis that progresses gradually. Advanced in vitro systems that recapitulate patient-like pathophysiology are emerging as alternatives to conventional animal-based models. In this review, we explore the interconnected pathogenic features of different types of ND, discuss the general strategy to modelling NDs using a microfluidic chip, and introduce the organoid-on-a-chip as the next advanced relevant model. Lastly, we overview how these models are being applied in academic and industrial drug development. The integration of microfluidic chips, stem cells, and biotechnological devices promises to provide valuable insights for biomedical research and developing diagnostic and therapeutic solutions for NDs.
Collapse
Affiliation(s)
- Sarnai Amartumur
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Huong Nguyen
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Thuy Huynh
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Testaverde S Kim
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea
| | - Ran-Sook Woo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon, 34824, Korea
| | - Eungseok Oh
- Department of Neurology, Chungnam National University Hospital, Daejeon, 35015, Korea
| | - Kyeong Kyu Kim
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Anti-microbial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea
| | - Luke P Lee
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Harvard Medical School, Division of Engineering in Medicine and Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA.
- Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, 94720, USA.
| | - Chaejeong Heo
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea.
| |
Collapse
|
42
|
Yildirim-Balatan C, Fenyi A, Besnault P, Gomez L, Sepulveda-Diaz JE, Michel PP, Melki R, Hunot S. Parkinson's disease-derived α-synuclein assemblies combined with chronic-type inflammatory cues promote a neurotoxic microglial phenotype. J Neuroinflammation 2024; 21:54. [PMID: 38383421 PMCID: PMC10882738 DOI: 10.1186/s12974-024-03043-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/12/2024] [Indexed: 02/23/2024] Open
Abstract
Parkinson's disease (PD) is a common age-related neurodegenerative disorder characterized by the aggregation of α-Synuclein (αSYN) building up intraneuronal inclusions termed Lewy pathology. Mounting evidence suggests that neuron-released αSYN aggregates could be central to microglial activation, which in turn mounts and orchestrates neuroinflammatory processes potentially harmful to neurons. Therefore, understanding the mechanisms that drive microglial cell activation, polarization and function in PD might have important therapeutic implications. Here, using primary microglia, we investigated the inflammatory potential of pure αSYN fibrils derived from PD patients. We further explored and characterized microglial cell responses to a chronic-type inflammatory stimulation combining PD patient-derived αSYN fibrils (FPD), Tumor necrosis factor-α (TNFα) and prostaglandin E2 (PGE2) (TPFPD). We showed that FPD hold stronger inflammatory potency than pure αSYN fibrils generated de novo. When combined with TNFα and PGE2, FPD polarizes microglia toward a particular functional phenotype departing from FPD-treated cells and featuring lower inflammatory cytokine and higher glutamate release. Whereas metabolomic studies showed that TPFPD-exposed microglia were closely related to classically activated M1 proinflammatory cells, notably with similar tricarboxylic acid cycle disruption, transcriptomic analysis revealed that TPFPD-activated microglia assume a unique molecular signature highlighting upregulation of genes involved in glutathione and iron metabolisms. In particular, TPFPD-specific upregulation of Slc7a11 (which encodes the cystine-glutamate antiporter xCT) was consistent with the increased glutamate response and cytotoxic activity of these cells toward midbrain dopaminergic neurons in vitro. Together, these data further extend the structure-pathological relationship of αSYN fibrillar polymorphs to their innate immune properties and demonstrate that PD-derived αSYN fibrils, TNFα and PGE2 act in concert to drive microglial cell activation toward a specific and highly neurotoxic chronic-type inflammatory phenotype characterized by robust glutamate release and iron retention.
Collapse
Affiliation(s)
- Cansu Yildirim-Balatan
- Sorbonne Université, Paris, France
- Institut du Cerveau - Paris Brain Institute - ICM, Hôpital de la Pitié-Salpêtrière, 91 Bd de l'Hôpital, 75013, Paris, France
- Inserm UMRS 1127, Paris, France
- CNRS UMR 7225, Paris, France
| | - Alexis Fenyi
- CEA and Laboratory of Neurodegenerative Diseases, CNRS, Institut François Jacob, MIRCen, 92265, Fontenay-aux-Roses, France
| | - Pierre Besnault
- Sorbonne Université, Paris, France
- Institut du Cerveau - Paris Brain Institute - ICM, Hôpital de la Pitié-Salpêtrière, 91 Bd de l'Hôpital, 75013, Paris, France
- Inserm UMRS 1127, Paris, France
- CNRS UMR 7225, Paris, France
| | - Lina Gomez
- Sorbonne Université, Paris, France
- Institut du Cerveau - Paris Brain Institute - ICM, Hôpital de la Pitié-Salpêtrière, 91 Bd de l'Hôpital, 75013, Paris, France
- Inserm UMRS 1127, Paris, France
- CNRS UMR 7225, Paris, France
| | - Julia E Sepulveda-Diaz
- Sorbonne Université, Paris, France
- Institut du Cerveau - Paris Brain Institute - ICM, Hôpital de la Pitié-Salpêtrière, 91 Bd de l'Hôpital, 75013, Paris, France
- Inserm UMRS 1127, Paris, France
- CNRS UMR 7225, Paris, France
| | - Patrick P Michel
- Sorbonne Université, Paris, France
- Institut du Cerveau - Paris Brain Institute - ICM, Hôpital de la Pitié-Salpêtrière, 91 Bd de l'Hôpital, 75013, Paris, France
- Inserm UMRS 1127, Paris, France
- CNRS UMR 7225, Paris, France
| | - Ronald Melki
- CEA and Laboratory of Neurodegenerative Diseases, CNRS, Institut François Jacob, MIRCen, 92265, Fontenay-aux-Roses, France
| | - Stéphane Hunot
- Sorbonne Université, Paris, France.
- Institut du Cerveau - Paris Brain Institute - ICM, Hôpital de la Pitié-Salpêtrière, 91 Bd de l'Hôpital, 75013, Paris, France.
- Inserm UMRS 1127, Paris, France.
- CNRS UMR 7225, Paris, France.
| |
Collapse
|
43
|
Shang G, Niu X, Tong Q, Zhao Y, Yin J, Zhou X, Xu J, Cao Y, Cheng F, Bao B, Li Z, Yao W. Integrated metabolomic and lipidomic analysis revealed the protective mechanisms of Erzhi Wan on senescent NRK cells through BRL cells. JOURNAL OF ETHNOPHARMACOLOGY 2024; 320:117482. [PMID: 38000520 DOI: 10.1016/j.jep.2023.117482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 11/08/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Erzhi Wan (EZW), as a prescription of traditional Chinese medicine, has been used for tonifying the liver and kidney. Although past studies have shown that EZW has potential anti-aging effect, the mechanisms associated with cellular metabolomics and lipidomics are not fully understood. AIM OF THE STUDY This study aimed to evaluate the anti-aging effect of EZW and investigate the mechanisms associated with cellular metabolomics and lipidomics. MATERIALS AND METHODS EZW solution at dosage of 3.6 g/kg in Sprague-Dawley rats was orally administered twice a day for 7 days and serum containing EZW was then collected. NRK cell senescence model induced by D-galactose was established in vitro, and non-contact co-culture cell assay was performed between senescent NRK cells and BRL cells intervened by serum containing EZW. The anti-aging effect of EZW on NRK cells was evaluated by metabolites identification, differential metabolites screening and metabolic pathways analysis through cellular metabolomics with GC-MS and lipidomics with UHPLC-Q-Exactive Orbitrap/MS. RESULTS Serum containing EZW indicated a protective effect through intervening BRL cells in non-contact co-culture system with D-gal-induced senescent NRK cells. For metabolic profiles, 71 endogenous metabolites were identified, among which 24 significantly differential metabolites were screened as metabolomics potential biomarkers. For lipidic profiles, 64 lipid components were identified in NRK cell samples under positive ion mode, among which 24 potential biomarkers of lipids were screened, mainly including PC and PE. 127 lipid components were identified in NRK cell samples under negative ion mode, among which 59 potential biomarkers of lipids were screened, including FA, PC, PE, PI and PS. Metabolic pathway analysis demonstrated that the identified differential metabolites found mainly involved in amino acids metabolism, energy metabolism and phospholipid biosynthesis pathways. CONCLUSION Serum containing EZW exhibited protective effect on D-gal-induced senescent NRK cells through intervening BRL cells by mainly regulating amino acids metabolism, energy metabolism and phospholipid biosynthesis pathways to possess its anti-aging function, providing a theoretical basis for clinical treatment of EZW.
Collapse
Affiliation(s)
- Guanxiong Shang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xuan Niu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Qingheng Tong
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yan Zhao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Jiu Yin
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xiaoqi Zhou
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Jia Xu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yudan Cao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Fangfang Cheng
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Beihua Bao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Zhipeng Li
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, 210009, China.
| | - Weifeng Yao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
44
|
Gupta S, Khan J, Ghosh S. Molecular mechanism of cognitive impairment associated with Parkinson's disease: A stroke perspective. Life Sci 2024; 337:122358. [PMID: 38128756 DOI: 10.1016/j.lfs.2023.122358] [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: 06/20/2023] [Revised: 12/03/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
Parkinson's disease (PD) is a common neurological illness that causes several motor and non-motor symptoms, most characteristically limb tremors and bradykinesia. PD is a slowly worsening disease that arises due to progressive neurodegeneration of specific areas of the brain, especially the substantia nigra of the midbrain. Even though PD has continuously been linked to a higher mortality risk in numerous epidemiologic studies, there have been significant discoveries regarding the connection between PD and stroke. The incidence of strokes such as cerebral infarction and hemorrhage is substantially associated with the development of PD. Moreover, cognitive impairments, primarily dementia, have been associated with stroke and PD. However, the underlying molecular mechanism of this phenomenon is still obscure. This concise review focuses on the relationship between stroke and PD, emphasizing the molecular mechanism of cognition deficit and memory loss evident in PD and stroke. Furthermore, we are also highlighting some potential drug molecules that can target both PD and stroke.
Collapse
Affiliation(s)
- Sanju Gupta
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur (IIT-Jodhpur), Rajasthan 342037, India
| | - Juhee Khan
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur (IIT-Jodhpur), Rajasthan 342037, India
| | - Surajit Ghosh
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur (IIT-Jodhpur), Rajasthan 342037, India.
| |
Collapse
|
45
|
Ratan Y, Rajput A, Pareek A, Pareek A, Jain V, Sonia S, Farooqui Z, Kaur R, Singh G. Advancements in Genetic and Biochemical Insights: Unraveling the Etiopathogenesis of Neurodegeneration in Parkinson's Disease. Biomolecules 2024; 14:73. [PMID: 38254673 PMCID: PMC10813470 DOI: 10.3390/biom14010073] [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: 11/21/2023] [Revised: 12/15/2023] [Accepted: 12/28/2023] [Indexed: 01/24/2024] Open
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative movement disorder worldwide, which is primarily characterized by motor impairments. Even though multiple hypotheses have been proposed over the decades that explain the pathogenesis of PD, presently, there are no cures or promising preventive therapies for PD. This could be attributed to the intricate pathophysiology of PD and the poorly understood molecular mechanism. To address these challenges comprehensively, a thorough disease model is imperative for a nuanced understanding of PD's underlying pathogenic mechanisms. This review offers a detailed analysis of the current state of knowledge regarding the molecular mechanisms underlying the pathogenesis of PD, with a particular emphasis on the roles played by gene-based factors in the disease's development and progression. This study includes an extensive discussion of the proteins and mutations of primary genes that are linked to PD, including α-synuclein, GBA1, LRRK2, VPS35, PINK1, DJ-1, and Parkin. Further, this review explores plausible mechanisms for DAergic neural loss, non-motor and non-dopaminergic pathologies, and the risk factors associated with PD. The present study will encourage the related research fields to understand better and analyze the current status of the biochemical mechanisms of PD, which might contribute to the design and development of efficacious and safe treatment strategies for PD in future endeavors.
Collapse
Affiliation(s)
- Yashumati Ratan
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India; (A.R.); (A.P.); (A.P.)
| | - Aishwarya Rajput
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India; (A.R.); (A.P.); (A.P.)
| | - Ashutosh Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India; (A.R.); (A.P.); (A.P.)
| | - Aaushi Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India; (A.R.); (A.P.); (A.P.)
| | - Vivek Jain
- Department of Pharmaceutical Sciences, Mohan Lal Sukhadia University, Udaipur 313001, Rajasthan, India;
| | - Sonia Sonia
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143005, Punjab, India;
| | - Zeba Farooqui
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL 60607, USA;
| | - Ranjeet Kaur
- Adesh Institute of Dental Sciences and Research, Bathinda 151101, Punjab, India;
| | - Gurjit Singh
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL 60607, USA;
| |
Collapse
|
46
|
Alborghetti M, Bianchini E, De Carolis L, Galli S, Pontieri FE, Rinaldi D. Type-B monoamine oxidase inhibitors in neurological diseases: clinical applications based on preclinical findings. Neural Regen Res 2024; 19:16-21. [PMID: 37488838 PMCID: PMC10479837 DOI: 10.4103/1673-5374.375299] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/06/2023] [Accepted: 05/04/2023] [Indexed: 07/26/2023] Open
Abstract
Type-B monoamine oxidase inhibitors, encompassing selegiline, rasagiline, and safinamide, are available to treat Parkinson's disease. These drugs ameliorate motor symptoms and improve motor fluctuation in the advanced stages of the disease. There is also evidence supporting the benefit of type-B monoamine oxidase inhibitors on non-motor symptoms of Parkinson's disease, such as mood deflection, cognitive impairment, sleep disturbances, and fatigue. Preclinical studies indicate that type-B monoamine oxidase inhibitors hold a strong neuroprotective potential in Parkinson's disease and other neurodegenerative diseases for reducing oxidative stress and stimulating the production and release of neurotrophic factors, particularly glial cell line-derived neurotrophic factor, which support dopaminergic neurons. Besides, safinamide may interfere with neurodegenerative mechanisms, counteracting excessive glutamate overdrive in basal ganglia motor circuit and reducing death from excitotoxicity. Due to the dual mechanism of action, the new generation of type-B monoamine oxidase inhibitors, including safinamide, is gaining interest in other neurological pathologies, and many supporting preclinical studies are now available. The potential fields of application concern epilepsy, Duchenne muscular dystrophy, multiple sclerosis, and above all, ischemic brain injury. The purpose of this review is to investigate the preclinical and clinical pharmacology of selegiline, rasagiline, and safinamide in Parkinson's disease and beyond, focusing on possible future therapeutic applications.
Collapse
Affiliation(s)
- Marika Alborghetti
- Neurology Unit, NESMOS Department, Faculty of Medicine & Psychology, Sapienza—University of Rome, Sant’Andrea University Hospital, Rome, Italy
| | - Edoardo Bianchini
- Neurology Unit, NESMOS Department, Faculty of Medicine & Psychology, Sapienza—University of Rome, Sant’Andrea University Hospital, Rome, Italy
- Department of Clinical and Behavioral Neurology, IRCCS—Fondazione Santa Lucia, Rome, Italy
| | - Lanfranco De Carolis
- Neurology Unit, NESMOS Department, Faculty of Medicine & Psychology, Sapienza—University of Rome, Sant’Andrea University Hospital, Rome, Italy
| | - Silvia Galli
- Neurology Unit, NESMOS Department, Faculty of Medicine & Psychology, Sapienza—University of Rome, Sant’Andrea University Hospital, Rome, Italy
| | - Francesco E. Pontieri
- Neurology Unit, NESMOS Department, Faculty of Medicine & Psychology, Sapienza—University of Rome, Sant’Andrea University Hospital, Rome, Italy
- Department of Clinical and Behavioral Neurology, IRCCS—Fondazione Santa Lucia, Rome, Italy
| | - Domiziana Rinaldi
- Neurology Unit, NESMOS Department, Faculty of Medicine & Psychology, Sapienza—University of Rome, Sant’Andrea University Hospital, Rome, Italy
- Department of Clinical and Behavioral Neurology, IRCCS—Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
47
|
Weaver MG, Popescu GK. Estimating the Ca 2+ Permeability of NMDA Receptors with Whole-Cell Patch-Clamp Electrophysiology. Methods Mol Biol 2024; 2799:177-200. [PMID: 38727908 PMCID: PMC11949104 DOI: 10.1007/978-1-0716-3830-9_10] [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: 07/03/2024]
Abstract
In the mammalian central nervous system (CNS), fast excitatory transmission relies primarily on the ionic fluxes generated by ionotropic glutamate receptors (iGluRs). Among iGluRs, NMDA receptors (NMDARs) are unique in their ability to pass large, Ca2+-rich currents. Importantly, their high Ca2+ permeability is essential for normal CNS function and is under physiological control. For this reason, the accurate measurement of NMDA receptor Ca2+ permeability represents a valuable experimental step in evaluating the mechanism by which these receptors contribute to a variety of physiological and pathological conditions. In this chapter, we provide a theoretical and practical overview of the common methods used to estimate the Ca2+ permeability of ion channels as they apply to NMDA receptors. Specifically, we describe the principles and methodology used to calculate relative permeability (PCa/PNa) and fractional permeability (Pf), along with the relationship between these two metrics. With increasing knowledge about the structural dynamics of ion channels and of the ongoing environmental fluctuations in which channels operate in vivo, the ability to quantify the Ca2+ entering cells through specific ion channels remains a tool essential to delineating the molecular mechanisms that support health and cause disease.
Collapse
Affiliation(s)
- Mae G Weaver
- Department of Biochemistry, Graduate Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, SUNY, Buffalo, NY, USA.
| | - Gabriela K Popescu
- Department of Biochemistry, Graduate Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, SUNY, Buffalo, NY, USA.
| |
Collapse
|
48
|
Vincent B, Shukla M. The Common Denominators of Parkinson's Disease Pathogenesis and Methamphetamine Abuse. Curr Neuropharmacol 2024; 22:2113-2156. [PMID: 37691228 PMCID: PMC11337683 DOI: 10.2174/1570159x21666230907151226] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 09/12/2023] Open
Abstract
The pervasiveness and mortality associated with methamphetamine abuse have doubled during the past decade, suggesting a possible worldwide substance use crisis. Epitomizing the pathophysiology and toxicology of methamphetamine abuse proclaims severe signs and symptoms of neurotoxic and neurobehavioral manifestations in both humans and animals. Most importantly, chronic use of this drug enhances the probability of developing neurodegenerative diseases manifolds. Parkinson's disease is one such neurological disorder, which significantly and evidently not only shares a number of toxic pathogenic mechanisms induced by methamphetamine exposure but is also interlinked both structurally and genetically. Methamphetamine-induced neurodegeneration involves altered dopamine homeostasis that promotes the aggregation of α-synuclein protofibrils in the dopaminergic neurons and drives these neurons to make them more vulnerable to degeneration, as recognized in Parkinson's disease. Moreover, the pathologic mechanisms such as mitochondrial dysfunction, oxidative stress, neuroinflammation and decreased neurogenesis detected in methamphetamine abusers dramatically resemble to what is observed in Parkinson's disease cases. Therefore, the present review comprehensively cumulates a holistic illustration of various genetic and molecular mechanisms putting across the notion of how methamphetamine administration and intoxication might lead to Parkinson's disease-like pathology and Parkinsonism.
Collapse
Affiliation(s)
- Bruno Vincent
- Institute of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560, Valbonne, France
| | - Mayuri Shukla
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, 10210, Bangkok, Thailand
| |
Collapse
|
49
|
Saramowicz K, Siwecka N, Galita G, Kucharska-Lusina A, Rozpędek-Kamińska W, Majsterek I. Alpha-Synuclein Contribution to Neuronal and Glial Damage in Parkinson's Disease. Int J Mol Sci 2023; 25:360. [PMID: 38203531 PMCID: PMC10778752 DOI: 10.3390/ijms25010360] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disease characterized by the progressive loss of dopaminergic neurons in the substantia nigra and the widespread accumulation of alpha-synuclein (αSyn) protein aggregates. αSyn aggregation disrupts critical cellular processes, including synaptic function, mitochondrial integrity, and proteostasis, which culminate in neuronal cell death. Importantly, αSyn pathology extends beyond neurons-it also encompasses spreading throughout the neuronal environment and internalization by microglia and astrocytes. Once internalized, glia can act as neuroprotective scavengers, which limit the spread of αSyn. However, they can also become reactive, thereby contributing to neuroinflammation and the progression of PD. Recent advances in αSyn research have enabled the molecular diagnosis of PD and accelerated the development of targeted therapies. Nevertheless, despite more than two decades of research, the cellular function, aggregation mechanisms, and induction of cellular damage by αSyn remain incompletely understood. Unraveling the interplay between αSyn, neurons, and glia may provide insights into disease initiation and progression, which may bring us closer to exploring new effective therapeutic strategies. Herein, we provide an overview of recent studies emphasizing the multifaceted nature of αSyn and its impact on both neuron and glial cell damage.
Collapse
Affiliation(s)
| | | | | | | | | | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (K.S.); (N.S.); (G.G.); (A.K.-L.); (W.R.-K.)
| |
Collapse
|
50
|
Remya C, Variyar EJ, Omkumar RV, Sadasivan C, Dileep KV. Unveiling the molecular basis of lobeline's allosteric regulation of NMDAR: insights from molecular modeling. Sci Rep 2023; 13:22418. [PMID: 38104236 PMCID: PMC10725453 DOI: 10.1038/s41598-023-49835-2] [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: 09/28/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023] Open
Abstract
Neurological and psychiatric disorders contribute significantly to the global disease burden, adversely affecting the quality of life for both patients and their families. Impaired glutamatergic signaling is considered to be a major cause for most of the neurological and psychiatric disorders. Glutamate receptors are over activated in excitotoxic conditions, leading to dysregulation of Ca2+ homeostasis, triggering the production of free radicals and oxidative stress, mitochondrial dysfunction and eventually cell death. Excitotoxicity primarily results from the overactivity of NMDARs, a subtype of ionotropic glutamate receptors, due to their pronounced Ca2+ permeability and conductance characteristics. NMDAR antagonists are suggested to have therapeutic use as they can prevent excitotoxicity. Our previous studies demonstrated lobeline, an alkaloid, exerts neuroprotective action in excitotoxic conditions by blocking NMDAR. However, the atomic level interactions of lobeline with NMDAR was not characterized yet. Structural comparison of lobeline with a known NMDAR antagonist ifenprodil, followed by molecular docking and dynamics simulations revealed that lobeline could bind to the ifenprodil binding site i.e., in the heterodimer interface of GluN1-GluN2B subunits and exert ifenprodil like activities. By in silico structure guided modifications on lobeline and subsequent free energy calculations, we propose putative NMDAR antagonists derived from lobeline.
Collapse
Affiliation(s)
- Chandran Remya
- Laboratory for Computational and Structural Biology, Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur, Kerala, 680005, India
| | - E J Variyar
- Department of Biotechnology and Microbiology, Kannur University, Dr. Janaki Ammal Campus, Thalassery, Kerala, 670661, India
| | - R V Omkumar
- Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thycaud PO, Trivandrum, Kerala, 695014, India
| | - C Sadasivan
- Department of Biotechnology and Microbiology, Kannur University, Dr. Janaki Ammal Campus, Thalassery, Kerala, 670661, India
- Inter University Centre for Bioscience, Kannur University, Dr. Janaki Ammal Campus, Thalassery, Kerala, 670661, India
| | - K V Dileep
- Laboratory for Computational and Structural Biology, Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur, Kerala, 680005, India.
| |
Collapse
|