1
|
Zagare A, Sauter T, Barmpa K, Pacheco M, Krüger R, Schwamborn JC, Saraiva C. MIRO1 mutation leads to metabolic maladaptation resulting in Parkinson's disease-associated dopaminergic neuron loss. NPJ Syst Biol Appl 2025; 11:37. [PMID: 40246848 PMCID: PMC12006346 DOI: 10.1038/s41540-025-00509-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 03/16/2025] [Indexed: 04/19/2025] Open
Abstract
MIRO1 is a mitochondrial outer membrane protein important for mitochondrial distribution, dynamics and bioenergetics. Over the last decade, evidence has pointed to a link between MIRO1 and Parkinson's disease (PD) pathogenesis. Moreover, a heterozygous MIRO1 mutation (p.R272Q) was identified in a PD patient, from which an iPSC-derived midbrain organoid model was derived, showing MIRO1 mutant-dependent selective loss of dopaminergic neurons. Herein, we use patient-specific iPSC-derived midbrain organoids carrying the MIRO1 p.R272Q mutation to further explore the cellular and molecular mechanisms involved in dopaminergic neuron degeneration. Using single-cell RNA sequencing (scRNAseq) analysis and metabolic modeling we show that the MIRO1 p.R272Q mutation affects the dopaminergic neuron developmental path leading to metabolic deficits and disrupted neuron-astrocyte metabolic crosstalk, which might represent an important pathogenic mechanism leading to their loss.
Collapse
Affiliation(s)
- Alise Zagare
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 2, place de l'Université, L-4365, Esch-sur-Alzette, Luxembourg
| | - Thomas Sauter
- Systems Biology and Epigenetics Group, Department of Life Sciences and Medicine, University of Luxembourg, 2, place de l'Université, L-4365, Esch-sur-Alzette, Luxembourg
| | - Kyriaki Barmpa
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 2, place de l'Université, L-4365, Esch-sur-Alzette, Luxembourg
| | - Maria Pacheco
- Systems Biology and Epigenetics Group, Department of Life Sciences and Medicine, University of Luxembourg, 2, place de l'Université, L-4365, Esch-sur-Alzette, Luxembourg
| | - Rejko Krüger
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 2, place de l'Université, L-4365, Esch-sur-Alzette, Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), 1 A-B rue Thomas Edison, L-1445, Strassen, Luxembourg
- Parkinson Research Clinic, Centre Hospitalier de Luxembourg, 4, rue Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Jens Christian Schwamborn
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 2, place de l'Université, L-4365, Esch-sur-Alzette, Luxembourg.
| | - Claudia Saraiva
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 2, place de l'Université, L-4365, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
2
|
McHale-Owen H, Faller KME, Chaytow H, Gillingwater TH. Phosphoglycerate kinase 1 as a therapeutic target in neurological disease. Trends Mol Med 2025:S1471-4914(25)00059-0. [PMID: 40234116 DOI: 10.1016/j.molmed.2025.03.008] [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: 12/03/2024] [Revised: 02/04/2025] [Accepted: 03/21/2025] [Indexed: 04/17/2025]
Abstract
Phosphoglycerate kinase 1 (PGK1) is a highly conserved enzyme that catalyzes the initial ATP-producing step in glycolysis. Improving cellular energy production by increasing PGK1 activity may be beneficial in multiple neurological conditions where cell metabolism is dysregulated, including Parkinson's disease (PD) and motor neuron disease (MND). This review examines recent evidence that suggests increasing PGK1 activity may be beneficial in multiple neurological conditions and discusses the current challenges surrounding the development of PGK1-focused therapies. PGK1 has considerable therapeutic potential, but novel PGK1 activators are needed to maximize the benefit for patients.
Collapse
Affiliation(s)
- Harriet McHale-Owen
- Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK; Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK
| | - Kiterie M E Faller
- Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK; Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK; Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Helena Chaytow
- Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK; Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK
| | - Thomas H Gillingwater
- Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK; Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
3
|
Rahimi Darehbagh R, Khanmohammadi S, Rezaei N. The role of mitochondrial DNA variants and dysfunction in the pathogenesis and progression of multiple sclerosis. Mitochondrion 2025; 81:102002. [PMID: 39732186 DOI: 10.1016/j.mito.2024.102002] [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: 03/30/2024] [Revised: 08/10/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease that affects the central nervous system (CNS). The etiology of MS remains elusive, with a complex interplay of genetic and environmental factors contributing to its pathogenesis. Recent studies showed mitochondrial DNA (mtDNA) as a potential player in the development and progression of MS. These studies encompassed mtDNA variants, copy number variations, and haplogroups. This narrative review aims to synthesize the current understanding of the role of mtDNA's in MS. The findings of this review suggest that mtDNA may indeed play a role in the development and progression of MS. Several studies have reported an association between mtDNA variants and increased susceptibility to MS, while others have found a link between mtDNA copy number variations and disease severity. Furthermore, specific mtDNA haplogroups have been demonstrated to confer protection against MS. MtDNA alterations may make neurons and oligodendrocytes more susceptible to inflammatory and oxidative stress, causing demyelination and axonal degeneration in MS patients. In conclusion, this review underscores the potential significance of mtDNA in the pathogenesis of MS and highlights the need for further research to fully elucidate its role. A deeper understanding of mtDNA's involvement in MS may pave the way for the development of novel therapeutic strategies to combat this debilitating disease.
Collapse
Affiliation(s)
- Ramyar Rahimi Darehbagh
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran; Nanoclub Elites Association, Tehran, Iran; Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran; Universal Scientific Education and Research Network (USERN), Sanandaj, Kurdistan, Iran
| | - Shaghayegh Khanmohammadi
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Zahoor I, Nematullah M, Ahmed ME, Fatma M, Sajad M, Ayasolla K, Cerghet M, Palaniyandi S, Ceci V, Carrera G, Buttari F, Centonze D, Mao-Draayer Y, Rattan R, Chiurchiù V, Giri S. Maresin-1 promotes neuroprotection and modulates metabolic and inflammatory responses in disease-associated cell types in preclinical models of multiple sclerosis. J Biol Chem 2025; 301:108226. [PMID: 39864620 PMCID: PMC11903811 DOI: 10.1016/j.jbc.2025.108226] [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: 07/23/2024] [Revised: 12/19/2024] [Accepted: 01/16/2025] [Indexed: 01/28/2025] Open
Abstract
Multiple sclerosis (MS) is a prevalent inflammatory neurodegenerative disease in young people, causing neurological abnormalities and impairment. To investigate a novel therapeutic agent for MS, we observed the impact of maresin 1 (MaR1) on disease progression in a well-known, relapsing-remitting experimental autoimmune encephalomyelitis mouse model. Treatment with MaR1 accelerated inflammation resolution, reduced neurological impairment, and delayed disease development by reducing immune cell infiltration (CD4+IL-17+ and CD4+IFNγ+) into the central nervous system. Furthermore, MaR1 administration enhanced IL-10 production, primarily in macrophages and CD4+ cells. However, neutralizing IL-10 with an anti-IL-10 antibody eliminated the protective impact by MaR1 in relapsing-remitting experimental autoimmune encephalomyelitis model, implying the significance of IL-10 in MaR1 treatment. Metabolism has been recognized as a critical mediator of effector activity in many types of immune cells. In our investigation, MaR1 administration significantly repaired metabolic dysregulation in CD4+ cells, macrophages, and microglia in EAE mice. Furthermore, MaR1 treatment restored defective efferocytosis in treated macrophages and microglia. MaR1 also preserved myelin in EAE mice and regulated O4+ oligodendrocyte metabolism by reversing metabolic dysregulation via increased mitochondrial activity and decreased glycolysis. Overall, in a preclinical MS animal model, MaR1 therapy has anti-inflammatory and neuroprotective properties. It also induced metabolic reprogramming in disease-associated cell types, increased efferocytosis, and maintained myelination. Moreover, our data on patient-derived peripheral blood mononuclear cells substantiated the protective role of MaR1, expanding the therapeutic spectrum of specialized proresolving lipid mediators. Altogether, these findings suggest the potential of MaR1 as a novel therapeutic agent for MS and other autoimmune diseases.
Collapse
MESH Headings
- Animals
- Docosahexaenoic Acids/pharmacology
- Docosahexaenoic Acids/therapeutic use
- Mice
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Multiple Sclerosis/drug therapy
- Multiple Sclerosis/metabolism
- Multiple Sclerosis/pathology
- Disease Models, Animal
- Female
- Inflammation/drug therapy
- Inflammation/metabolism
- Inflammation/pathology
- Macrophages/metabolism
- Macrophages/drug effects
- Macrophages/pathology
- Macrophages/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/pathology
- Humans
- Mice, Inbred C57BL
- Interleukin-10/metabolism
- Neuroprotection/drug effects
- Neuroprotective Agents/pharmacology
Collapse
Affiliation(s)
- Insha Zahoor
- Department of Neurology, Henry Ford Health, Detroit, Michigan, USA
| | | | | | - Mena Fatma
- Department of Neurology, Henry Ford Health, Detroit, Michigan, USA
| | - Mir Sajad
- Department of Neurology, Henry Ford Health, Detroit, Michigan, USA
| | | | - Mirela Cerghet
- Department of Neurology, Henry Ford Health, Detroit, Michigan, USA
| | - Suresh Palaniyandi
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health, Detroit, Michigan, USA; Department of Physiology, Wayne State University, Detroit, Michigan, USA
| | - Veronica Ceci
- Institute of Translational Pharmacology, National Research Council, Rome, Italy; Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Giulia Carrera
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Fabio Buttari
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), Italy
| | - Diego Centonze
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), Italy
| | - Yang Mao-Draayer
- Oklahoma Medical Research Foundation, Oklahoma, Farmington Hills, Michigan, USA
| | - Ramandeep Rattan
- Women's Health Services, Henry Ford Health, Detroit, Michigan, USA
| | - Valerio Chiurchiù
- Institute of Translational Pharmacology, National Research Council, Rome, Italy; Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Shailendra Giri
- Department of Neurology, Henry Ford Health, Detroit, Michigan, USA.
| |
Collapse
|
5
|
Osmanovic Barilar J, Papic V, Farkas V, Rubic I, Meglic P, Bagaric R, Babic Perhoc A, Virag D, Homolak J, Salkovic-Petrisic M, Knezovic A. Modeling of Parkinson's disease by intrastriatal administration of streptozotocin. Neuropharmacology 2025; 265:110246. [PMID: 39643239 DOI: 10.1016/j.neuropharm.2024.110246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/21/2024] [Accepted: 12/01/2024] [Indexed: 12/09/2024]
Abstract
Parkinson's disease (PD) is a highly heterogeneous and therefore a possible cause of translation failure of drugs from animal testing to human treatments can be because existing models cannot replicate the entire spectrum of PD features. One of the theories of the origin of neurodegenerative diseases assumes metabolic dysfunction as a common fundamental thread of disease development. Intracerebroventricular administration of streptozotocin induces insulin resistance in the brain (Alzheimer's disease animal model). The aim of this project is to examine whether metabolic dysfunction caused by direct application of streptozotocin to brain region affected in PD (striatum) can induce characteristic PD symptoms. Adult male Wistar rats were given streptozotocin bilaterally or unilaterally in striatum. PET scan, cognitive, behavioural and motoric functions were tested one month after administration. Metabolite and protein analysis was done by untargeted metabolomics, ELISA and Western blot. Rats administered bilaterally showed motoric deficit, cognitive deficit of spatial learning and memory, fear conditioned and recognition memory, and anxiety-like behaviour, accompanied by impaired brain glucose uptake and metabolism. The results provide first evidence that bilateral intrastriatal administration of streptozotocin (particularly lower dose) can cause development of the hallmark PD symptoms. As metabolic dysfunction is increasingly associated with PD, an animal model with hypermetabolism in the early-on could be a better PD model for testing diverse therapeutics and the results could be better translated to humans. Further characterization is needed for understanding possible underlying mechanism and development of a new animal model for unique PD endophenotype expressing motoric, cognitive and metabolic symptomatology.
Collapse
Affiliation(s)
- Jelena Osmanovic Barilar
- Department of Pharmacology, School of Medicine University of Zagreb, Croatia; Croatian Institute for Brain Research, School of Medicine University of Zagreb, Croatia
| | - Vito Papic
- Department of Pharmacology, School of Medicine University of Zagreb, Croatia; Croatian Institute for Brain Research, School of Medicine University of Zagreb, Croatia
| | - Vladimir Farkas
- Clinic for Internal Diseases, Faculty of Veterinary Medicine, University of Zagreb, Croatia
| | - Ivana Rubic
- Clinic for Internal Diseases, Faculty of Veterinary Medicine, University of Zagreb, Croatia
| | - Patrik Meglic
- Department of Pharmacology, School of Medicine University of Zagreb, Croatia; Croatian Institute for Brain Research, School of Medicine University of Zagreb, Croatia
| | - Robert Bagaric
- Department of Experimental Physics, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Ana Babic Perhoc
- Department of Pharmacology, School of Medicine University of Zagreb, Croatia; Croatian Institute for Brain Research, School of Medicine University of Zagreb, Croatia
| | - Davor Virag
- Department of Pharmacology, School of Medicine University of Zagreb, Croatia; Croatian Institute for Brain Research, School of Medicine University of Zagreb, Croatia
| | - Jan Homolak
- Department of Pharmacology, School of Medicine University of Zagreb, Croatia; Croatian Institute for Brain Research, School of Medicine University of Zagreb, Croatia; M3 Research Institute & Interfaculty Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen, Germany
| | - Melita Salkovic-Petrisic
- Department of Pharmacology, School of Medicine University of Zagreb, Croatia; Croatian Institute for Brain Research, School of Medicine University of Zagreb, Croatia
| | - Ana Knezovic
- Department of Pharmacology, School of Medicine University of Zagreb, Croatia; Croatian Institute for Brain Research, School of Medicine University of Zagreb, Croatia.
| |
Collapse
|
6
|
Lv R, Liu B, Jiang Z, Zhou R, Liu X, Lu T, Bao Y, Huang C, Zou G, Zhang Z, Lu L, Yin Q. Intermittent fasting and neurodegenerative diseases: Molecular mechanisms and therapeutic potential. Metabolism 2025; 164:156104. [PMID: 39674569 DOI: 10.1016/j.metabol.2024.156104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
Neurodegenerative disorders are straining public health worldwide. During neurodegenerative disease progression, aberrant neuronal network activity, bioenergetic impairment, adaptive neural plasticity impairment, dysregulation of neuronal Ca2+ homeostasis, oxidative stress, and immune inflammation manifest as characteristic pathological changes in the cellular milieu of the brain. There is no drug for the treatment of neurodegenerative disorders, and therefore, strategies/treatments for the prevention or treatment of neurodegenerative disorders are urgently needed. Intermittent fasting (IF) is characterized as an eating pattern that alternates between periods of fasting and eating, requiring fasting durations that vary depending on the specific protocol implemented. During IF, depletion of liver glycogen stores leads to the production of ketone bodies from fatty acids derived from adipocytes, thereby inducing an altered metabolic state accompanied by cellular and molecular adaptive responses within neural networks in the brain. At the cellular level, adaptive responses can promote the generation of synapses and neurons. At the molecular level, IF triggers the activation of associated transcription factors, thereby eliciting the expression of protective proteins. Consequently, this regulatory process governs central and peripheral metabolism, oxidative stress, inflammation, mitochondrial function, autophagy, and the gut microbiota, all of which contribute to the amelioration of neurodegenerative disorders. Emerging evidence suggests that weight regulation significantly contributes to the neuroprotective effects of IF. By alleviating obesity-related factors such as blood-brain barrier dysfunction, neuroinflammation, and β-amyloid accumulation, IF enhances metabolic flexibility and insulin sensitivity, further supporting its potential in mitigating neurodegenerative disorders. The present review summarizes animal and human studies investigating the role and underlying mechanisms of IF in physiology and pathology, with an emphasis on its therapeutic potential. Furthermore, we provide an overview of the cellular and molecular mechanisms involved in regulating brain energy metabolism through IF, highlighting its potential applications in neurodegenerative disorders. Ultimately, our findings offer novel insights into the preventive and therapeutic applications of IF for neurodegenerative disorders.
Collapse
Affiliation(s)
- Renjun Lv
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| | - Bin Liu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan 250014, China
| | - Ziying Jiang
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, 100053, China
| | - Runfa Zhou
- Experimental Pharmacology Mannheim, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehlstr. 13-17, Mannheim 68167, Germany
| | - Xiaoxing Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191 Beijing, China
| | - Tangsheng Lu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China
| | - Yanping Bao
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China
| | - Chunxia Huang
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China
| | - Guichang Zou
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China
| | - Zongyong Zhang
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China.
| | - Lin Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191 Beijing, China; National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, 100871 Beijing, China.
| | - Qingqing Yin
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| |
Collapse
|
7
|
Souder DC, McGregor ER, Clark JP, Rhoads TW, Porter TJ, Eliceiri KW, Moore DL, Puglielli L, Anderson RM. Neuron-specific isoform of PGC-1α regulates neuronal metabolism and brain aging. Nat Commun 2025; 16:2053. [PMID: 40021651 PMCID: PMC11871081 DOI: 10.1038/s41467-025-57363-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 02/19/2025] [Indexed: 03/03/2025] Open
Abstract
The brain is a high-energy tissue, and although aging is associated with dysfunctional inflammatory and neuron-specific functional pathways, a direct connection to metabolism is not established. Here, we show that isoforms of mitochondrial regulator PGC-1α are driven from distinct brain cell-type specific promotors, repressed with aging, and integral in coordinating metabolism and growth signaling. Transcriptional and proteomic profiles of cortex from male adult, middle age, and advanced age mice reveal an aging metabolic signature linked to PGC-1α. In primary culture, a neuron-exclusive promoter produces the functionally dominant isoform of PGC-1α. Using growth repression as a challenge, we find that PGC-1α is regulated downstream of GSK3β independently across promoters. Broad cellular metabolic consequences of growth inhibition observed in vitro are mirrored in vivo, including activation of PGC-1α directed programs and suppression of aging pathways. These data place PGC-1α centrally in a growth and metabolism network directly relevant to brain aging.
Collapse
Affiliation(s)
- Dylan C Souder
- Department of Medicine, SMPH, University of Wisconsin Madison, Madison, WI, USA
| | - Eric R McGregor
- Department of Medicine, SMPH, University of Wisconsin Madison, Madison, WI, USA
| | - Josef P Clark
- Department of Medicine, SMPH, University of Wisconsin Madison, Madison, WI, USA
| | - Timothy W Rhoads
- Department of Nutritional Sciences, University of Wisconsin Madison, Madison, WI, USA
| | - Tiaira J Porter
- Department of Neuroscience, University of Wisconsin Madison, Madison, WI, USA
| | - Kevin W Eliceiri
- Department of Medical Physics, University of Wisconsin Madison, Madison, WI, USA
| | - Darcie L Moore
- Department of Neuroscience, University of Wisconsin Madison, Madison, WI, USA
| | - Luigi Puglielli
- Department of Medicine, SMPH, University of Wisconsin Madison, Madison, WI, USA
- GRECC William S, Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Rozalyn M Anderson
- Department of Medicine, SMPH, University of Wisconsin Madison, Madison, WI, USA.
- GRECC William S, Middleton Memorial Veterans Hospital, Madison, WI, USA.
| |
Collapse
|
8
|
Ladjel-Mendil A, Ahras-Sifi N, Moussaoui H, Chérifi F, Laraba-Djebari F. Immunomodulatory effect of selective COX-2 inhibitor celecoxib on the neuropathological disorders and immunoinflammatory response induced by Kaliotoxin from Androctonus australis venom. Toxicon 2025; 255:108265. [PMID: 39884560 DOI: 10.1016/j.toxicon.2025.108265] [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: 09/14/2024] [Revised: 01/18/2025] [Accepted: 01/23/2025] [Indexed: 02/01/2025]
Abstract
The immune response is increasingly being linked to the pathogenic processes underlying neurological disorders including potassium channel malfunction. Few investigations, meanwhile, have shown how cyclooxygenase-2 (COX-2) is involved in the neuroimmunopathology linked to potassium channel failure. Thus, using an animal model of neuropathology caused by kaliotoxin, an exclusive blocker of voltage-gated potassium channels from the scorpion venom of Androctonus australis hector, we examined the immunomodulatory impact of celecoxib (selective inhibitor of COX-2). The neural and systemic pathogenic effects of KTX can be considerably reduced by celecoxib-mediated COX-2 inhibition, according to the results. It most certainly works via controlling the immunoinflammatory exposure by raising IL-10 levels; decreasing proinflammatory cytokine levels including mostly TNFα and IL-6, and balancing oxidative status. Along with that, by significantly promoting tissue healing, COX-2 inhibitor also enhances cellular metabolism. One potential treatment approach for immunoinflammatory exacerbations linked to neurodegenerative is the COX-2 inhibitor.
Collapse
Affiliation(s)
- Amina Ladjel-Mendil
- USTHB, Faculty of Biological Sciences, Laboratory of Cellular and Molecular Biology, BP 32, El-Alia Bab Ezzouar, 16111, Algiers, Algeria.
| | - Nesrine Ahras-Sifi
- USTHB, Faculty of Biological Sciences, Laboratory of Cellular and Molecular Biology, BP 32, El-Alia Bab Ezzouar, 16111, Algiers, Algeria.
| | - Hadjila Moussaoui
- USTHB, Faculty of Biological Sciences, Laboratory of Cellular and Molecular Biology, BP 32, El-Alia Bab Ezzouar, 16111, Algiers, Algeria.
| | - Fatah Chérifi
- USTHB, Faculty of Biological Sciences, Laboratory of Cellular and Molecular Biology, BP 32, El-Alia Bab Ezzouar, 16111, Algiers, Algeria.
| | - Fatima Laraba-Djebari
- USTHB, Faculty of Biological Sciences, Laboratory of Cellular and Molecular Biology, BP 32, El-Alia Bab Ezzouar, 16111, Algiers, Algeria; Algerian Academy of Sciences and Technology, Villa Rais Hamidou, Chemin Omar Kachkar, El Madania, Algiers, Algeria.
| |
Collapse
|
9
|
Breßer M, Siemens KD, Schneider L, Lunnebach JE, Leven P, Glowka TR, Oberländer K, De Domenico E, Schultze JL, Schmidt J, Kalff JC, Schneider A, Wehner S, Schneider R. Macrophage-induced enteric neurodegeneration leads to motility impairment during gut inflammation. EMBO Mol Med 2025; 17:301-335. [PMID: 39762650 PMCID: PMC11822118 DOI: 10.1038/s44321-024-00189-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 02/14/2025] Open
Abstract
Current studies pictured the enteric nervous system and macrophages as modulators of neuroimmune processes in the inflamed gut. Expanding this view, we investigated the impact of enteric neuron-macrophage interactions on postoperative trauma and subsequent motility disturbances, i.e., postoperative ileus. In the early postsurgical phase, we detected strong neuronal activation, followed by transcriptional and translational signatures indicating neuronal death and synaptic damage. Simultaneously, our study revealed neurodegenerative profiles in macrophage-specific transcriptomes after postoperative trauma. Validating the role of resident and monocyte-derived macrophages, we depleted macrophages by CSF-1R-antibodies and used CCR2-/- mice, known for reduced monocyte infiltration, in POI studies. Only CSF-1R-antibody-treated animals showed decreased neuronal death and lessened synaptic decay, emphasizing the significance of resident macrophages. In human gut samples taken early and late during abdominal surgery, we substantiated the mouse model data and found reactive and apoptotic neurons and dysregulation in synaptic genes, indicating a species' overarching mechanism. Our study demonstrates that surgical trauma activates enteric neurons and induces neurodegeneration, mediated by resident macrophages, introducing neuroprotection as an option for faster recovery after surgery.
Collapse
Affiliation(s)
- Mona Breßer
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Kevin D Siemens
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Linda Schneider
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | | | - Patrick Leven
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Tim R Glowka
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Kristin Oberländer
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University of Bonn Medical Center, Dept. of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, 53127, Bonn, Germany
| | - Elena De Domenico
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE). PRECISE Platform for Genomics and Epigenomics at DZNE and University of Bonn, Bonn, Germany
| | - Joachim L Schultze
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE). PRECISE Platform for Genomics and Epigenomics at DZNE and University of Bonn, Bonn, Germany
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Joachim Schmidt
- Department of General, Thoracic and Vascular Surgery, University Hospital Bonn, Bonn, Germany
| | - Jörg C Kalff
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University of Bonn Medical Center, Dept. of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, 53127, Bonn, Germany
| | - Sven Wehner
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | | |
Collapse
|
10
|
Gubin D, Weinert D, Stefani O, Otsuka K, Borisenkov M, Cornelissen G. Wearables in Chronomedicine and Interpretation of Circadian Health. Diagnostics (Basel) 2025; 15:327. [PMID: 39941257 PMCID: PMC11816745 DOI: 10.3390/diagnostics15030327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/14/2025] [Accepted: 01/26/2025] [Indexed: 02/16/2025] Open
Abstract
Wearable devices have gained increasing attention for use in multifunctional applications related to health monitoring, particularly in research of the circadian rhythms of cognitive functions and metabolic processes. In this comprehensive review, we encompass how wearables can be used to study circadian rhythms in health and disease. We highlight the importance of these rhythms as markers of health and well-being and as potential predictors for health outcomes. We focus on the use of wearable technologies in sleep research, circadian medicine, and chronomedicine beyond the circadian domain and emphasize actigraphy as a validated tool for monitoring sleep, activity, and light exposure. We discuss various mathematical methods currently used to analyze actigraphic data, such as parametric and non-parametric approaches, linear, non-linear, and neural network-based methods applied to quantify circadian and non-circadian variability. We also introduce novel actigraphy-derived markers, which can be used as personalized proxies of health status, assisting in discriminating between health and disease, offering insights into neurobehavioral and metabolic status. We discuss how lifestyle factors such as physical activity and light exposure can modulate brain functions and metabolic health. We emphasize the importance of establishing reference standards for actigraphic measures to further refine data interpretation and improve clinical and research outcomes. The review calls for further research to refine existing tools and methods, deepen our understanding of circadian health, and develop personalized healthcare strategies.
Collapse
Affiliation(s)
- Denis Gubin
- Department of Biology, Tyumen Medical University, 625023 Tyumen, Russia
- Laboratory for Chronobiology and Chronomedicine, Research Institute of Biomedicine and Biomedical Technologies, Tyumen Medical University, 625023 Tyumen, Russia
- Tyumen Cardiology Research Center, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
| | - Dietmar Weinert
- Institute of Biology/Zoology, Martin Luther University, 06108 Halle-Wittenberg, Germany;
| | - Oliver Stefani
- Department Engineering and Architecture, Institute of Building Technology and Energy, Lucerne University of Applied Sciences and Arts, 6048 Horw, Switzerland;
| | - Kuniaki Otsuka
- Tokyo Women’s Medical University, Tokyo 162-8666, Japan;
- Halberg Chronobiology Center, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Mikhail Borisenkov
- Department of Molecular Immunology and Biotechnology, Institute of Physiology of Komi Science Centre, Ural Branch of the Russian Academy of Sciences, 167982 Syktyvkar, Russia;
| | - Germaine Cornelissen
- Halberg Chronobiology Center, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
11
|
Apparoo Y, Phan CW, Kuppusamy UR, Wei Chiang EC. Ergothioneine-rich Lentinula edodes mushroom extract restores mitochondrial functions in senescent HT22 cells. Neuroscience 2025; 565:277-291. [PMID: 39643233 DOI: 10.1016/j.neuroscience.2024.11.082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 11/21/2024] [Accepted: 11/30/2024] [Indexed: 12/09/2024]
Abstract
A decline in mitochondrial functions associated with ageing is the key factor of free radical generation which contributes to age-related pathologies. Protecting healthy functional mitochondrial networks with antioxidants is critical in promoting healthy ageing. This study aimed to investigate the protective effect of ergothioneine (EGT)-rich Lentinula edodes extract (LE-ETH) against tert-butyl hydroperoxide (t-BHP) assaulted senescent HT22 cells. Mitochondrial function was evaluated by measuring mitochondrial membrane potential (MMP), ATP levels and mitochondrial toxicity. The protective mechanisms were elucidated via the exploration of antioxidant and mitochondrial biogenesis signalling pathways. Our results revealed that a low dose of t-BHP increases mitochondrial toxicity. The pretreatment with 100 µg/mL of LE-ETH and the equimolar concentration of EGT for 8 h significantly improve the mitochondrial function and reduced inflammation. Through gene expression studies, we demonstrated that pretreatment of LE-ETH significantly improves the antioxidant and mitochondrial biogenesis pathway via Nrf2 signaling axis. However, the downstream genes of the mitochondrial biogenesis pathway were unaffected by equimolar EGT concentration. Gas chromatography-mass spectrum (GC-MS) analysis was carried out to identify the bioactive compounds that are present in LE-ETH extract which contributed to its efficacy in improving the mitochondrial functions. A total of 23 compounds consisting of phenols, fatty acids, and sterols were identified in the ethanolic extract. Pentanoic acid was the major compound identified in LE-ETH. These findings demonstrated that EGT-rich L.edodes mushroom is a potential neuroprotective agent which could serve as a potential therapeutic strategy for the preservation of mitochondrial functions in healthy ageing explorations.
Collapse
Affiliation(s)
- Yasaaswini Apparoo
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Chia Wei Phan
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Malaya, 50603 Kuala Lumpur, Malaysia; Neuroscience Research Group (NeuRG), Faculty of Pharmacy, Universiti Malaya, 50603 Kuala Lumpur, Malaysia; Mushroom Research Centre, Universiti Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Umah Rani Kuppusamy
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Eric Chan Wei Chiang
- Department of Food Science with Nutrition, Faculty of Applied Sciences, UCSI University, 56000, Cheras, Kuala Lumpur, Malaysia
| |
Collapse
|
12
|
Szablewski L. Associations Between Diabetes Mellitus and Neurodegenerative Diseases. Int J Mol Sci 2025; 26:542. [PMID: 39859258 PMCID: PMC11765393 DOI: 10.3390/ijms26020542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Diabetes mellitus (DM) and neurodegenerative diseases/disturbances are worldwide health problems. The most common chronic conditions diagnosed in persons 60 years and older are type 2 diabetes mellitus (T2DM) and cognitive impairment. It was found that diabetes mellitus is a major risk for cognitive decline, dementia, Parkinson's disease (PD), Alzheimer's disease (AD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS) and other neurodegenerative disorders. Different mechanisms of associations between these diseases and diabetes mellitus have been suggested. For example, it is postulated that an impaired intracellular insulin signaling pathway, together with hyperglycemia and hyperinsulinemia, may cause pathological changes, such as dysfunction of the mitochondria, oxidative stress inflammatory responses, etc. The association between diabetes mellitus and neurodegenerative diseases, as well as the mechanisms of these associations, needs further investigation. The aim of this review is to describe the associations between diabetes mellitus, especially type 1 (T1DM) and type 2 diabetes mellitus, and selected neurodegenerative diseases, i.e., Alzheimer's disease, Parkinson's disease, Huntington's disease and amyotrophic lateral sclerosis. Suggested mechanisms of these associations are also described.
Collapse
Affiliation(s)
- Leszek Szablewski
- Chair and Department of General Biology and Parasitology, Medical University of Warsaw, Chałubińskiego 5, 02-004 Warsaw, Poland
| |
Collapse
|
13
|
Deng YT, You J, He Y, Zhang Y, Li HY, Wu XR, Cheng JY, Guo Y, Long ZW, Chen YL, Li ZY, Yang L, Zhang YR, Chen SD, Ge YJ, Huang YY, Shi LM, Dong Q, Mao Y, Feng JF, Cheng W, Yu JT. Atlas of the plasma proteome in health and disease in 53,026 adults. Cell 2025; 188:253-271.e7. [PMID: 39579765 DOI: 10.1016/j.cell.2024.10.045] [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: 03/24/2024] [Revised: 07/17/2024] [Accepted: 10/24/2024] [Indexed: 11/25/2024]
Abstract
Large-scale proteomics studies can refine our understanding of health and disease and enable precision medicine. Here, we provide a detailed atlas of 2,920 plasma proteins linking to diseases (406 prevalent and 660 incident) and 986 health-related traits in 53,026 individuals (median follow-up: 14.8 years) from the UK Biobank, representing the most comprehensive proteome profiles to date. This atlas revealed 168,100 protein-disease associations and 554,488 protein-trait associations. Over 650 proteins were shared among at least 50 diseases, and over 1,000 showed sex and age heterogeneity. Furthermore, proteins demonstrated promising potential in disease discrimination (area under the curve [AUC] > 0.80 in 183 diseases). Finally, integrating protein quantitative trait locus data determined 474 causal proteins, providing 37 drug-repurposing opportunities and 26 promising targets with favorable safety profiles. These results provide an open-access comprehensive proteome-phenome resource (https://proteome-phenome-atlas.com/) to help elucidate the biological mechanisms of diseases and accelerate the development of disease biomarkers, prediction models, and therapeutic targets.
Collapse
Affiliation(s)
- Yue-Ting Deng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jia You
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China; Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Yu He
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hai-Yun Li
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xin-Rui Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ji-Yun Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Guo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zi-Wen Long
- Department of Gastric Cancer Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Lin Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ze-Yu Li
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China; Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Liu Yang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ya-Ru Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Jun Ge
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Yuan Huang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Le-Ming Shi
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital Fudan University, Shanghai, China.
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China; Department of Computer Science, University of Warwick, Coventry, UK.
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China; Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
14
|
Canovai A, Williams PA. Pyrroloquinoline quinone: a potential neuroprotective compound for neurodegenerative diseases targeting metabolism. Neural Regen Res 2025; 20:41-53. [PMID: 38767475 PMCID: PMC11246121 DOI: 10.4103/nrr.nrr-d-23-01921] [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: 11/23/2023] [Revised: 01/31/2024] [Accepted: 02/29/2024] [Indexed: 05/22/2024] Open
Abstract
Pyrroloquinoline quinone is a quinone described as a cofactor for many bacterial dehydrogenases and is reported to exert an effect on metabolism in mammalian cells/tissues. Pyrroloquinoline quinone is present in the diet being available in foodstuffs, conferring the potential of this compound to be supplemented by dietary administration. Pyrroloquinoline quinone's nutritional role in mammalian health is supported by the extensive deficits in reproduction, growth, and immunity resulting from the dietary absence of pyrroloquinoline quinone, and as such, pyrroloquinoline quinone has been considered as a "new vitamin." Although the classification of pyrroloquinoline quinone as a vitamin needs to be properly established, the wide range of benefits for health provided has been reported in many studies. In this respect, pyrroloquinoline quinone seems to be particularly involved in regulating cell signaling pathways that promote metabolic and mitochondrial processes in many experimental contexts, thus dictating the rationale to consider pyrroloquinoline quinone as a vital compound for mammalian life. Through the regulation of different metabolic mechanisms, pyrroloquinoline quinone may improve clinical deficits where dysfunctional metabolism and mitochondrial activity contribute to induce cell damage and death. Pyrroloquinoline quinone has been demonstrated to have neuroprotective properties in different experimental models of neurodegeneration, although the link between pyrroloquinoline quinone-promoted metabolism and improved neuronal viability in some of such contexts is still to be fully elucidated. Here, we review the general properties of pyrroloquinoline quinone and its capacity to modulate metabolic and mitochondrial mechanisms in physiological contexts. In addition, we analyze the neuroprotective properties of pyrroloquinoline quinone in different neurodegenerative conditions and consider future perspectives for pyrroloquinoline quinone's potential in health and disease.
Collapse
Affiliation(s)
| | - Pete A. Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
15
|
Pluta R, Kocki J, Bogucki J, Bogucka-Kocka A, Czuczwar SJ. Apolipoprotein ( APOA1, APOE, CLU) genes expression in the CA3 region of the hippocampus in an ischemic model of Alzheimer's disease with survival up to 2 years. J Alzheimers Dis 2025; 103:627-634. [PMID: 39686609 DOI: 10.1177/13872877241303950] [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: 12/18/2024]
Abstract
BACKGROUND Changes in the Alzheimer's disease-related apolipoprotein genes expression, occurring parallel with brain ischemia-induced neurodegeneration in the hippocampal CA3 area, may be crucial for the development of memory loss and dementia. OBJECTIVE The aim of the study was to investigate changes in genes expression of apolipoprotein A1 (APOA1), apolipoprotein E (APOE), and clusterin (CLU) in CA3 area post-ischemia with survival of 2 years. METHODS The gene expression was evaluated with the use of an RT-PCR protocol after 2, 7, and 30 days and 6, 12, 18, and 24 months post-ischemia. RESULTS The expression of the APOA1 gene (encoding apolipoprotein A1) was below the control values at 2 days, 6 and 12 months while at 7 and 30 days and 18 and 24 months post-ischemia this gene expression exceeded the control values. In the case of the CLU gene (encoding clusterin) expression, it was above the control values at all times post-ischemia. Similar expression was observed for the APOE gene (encoding apolipoprotein E) except on day 7 after ischemia where its expression was below the control value. CONCLUSIONS The results seem to indicate that the observed changes in the gene expression may reflect the activation and inhibition of a variety of processes involved in ischemia-induced neurodegeneration. The enhanced expression of APOA1 and CLU genes may be associated with induction of neuroprotective mechanisms while increased expression of the APOE gene may produce detrimental effects.
Collapse
Affiliation(s)
- Ryszard Pluta
- Department of Pathophysiology, Medical University of Lublin, Lublin, Poland
| | - Janusz Kocki
- Department of Clinical Genetics, Medical University of Lublin, Lublin, Poland
| | - Jacek Bogucki
- Faculty of Medicine, John Paul II Catholic University of Lublin, Lublin, Poland
| | - Anna Bogucka-Kocka
- Department of Biology and Genetics, Medical University of Lublin, Lublin, Poland
| | | |
Collapse
|
16
|
Perna L, Salsbury G, Dushti M, Smith CJ, Morales V, Bianchi K, Czibik G, Chapple JP. Altered Cellular Metabolism Is a Consequence of Loss of the Ataxia-Linked Protein Sacsin. Int J Mol Sci 2024; 25:13242. [PMID: 39769008 PMCID: PMC11675909 DOI: 10.3390/ijms252413242] [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: 11/04/2024] [Revised: 11/28/2024] [Accepted: 12/03/2024] [Indexed: 01/30/2025] Open
Abstract
Mitochondrial dysfunction is implicated in the pathogenesis of the neurological condition autosomal recessive spastic ataxia of Charlevoix-Saguenay (ARSACS), yet precisely how the mitochondrial metabolism is affected is unknown. Thus, to better understand changes in the mitochondrial metabolism caused by loss of the sacsin protein (encoded by the SACS gene, which is mutated in ARSACS), we performed mass spectrometry-based tracer analysis, with both glucose- and glutamine-traced carbon. Comparing the metabolite profiles between wild-type and sacsin-knockout cell lines revealed increased reliance on aerobic glycolysis in sacsin-deficient cells, as evidenced by the increase in lactate and reduction of glucose. Moreover, sacsin knockout cells differentiated towards a neuronal phenotype had increased levels of tricarboxylic acid cycle metabolites relative to the controls. We also observed disruption in the glutaminolysis pathway in differentiated and undifferentiated cells in the absence of sacsin. In conclusion, this work demonstrates consequences for cellular metabolism associated with a loss of sacsin, which may be relevant to ARSACS.
Collapse
Affiliation(s)
- Laura Perna
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; (L.P.); (G.S.); (M.D.); (C.J.S.); (G.C.)
| | - Grace Salsbury
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; (L.P.); (G.S.); (M.D.); (C.J.S.); (G.C.)
| | - Mohammed Dushti
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; (L.P.); (G.S.); (M.D.); (C.J.S.); (G.C.)
- Pharmacology & Toxicology Department, Faculty of Medicine, Kuwait University, Kuwait City P.O. Box 24923, Kuwait
| | - Christopher J. Smith
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; (L.P.); (G.S.); (M.D.); (C.J.S.); (G.C.)
| | - Valle Morales
- Barts Cancer Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; (V.M.); (K.B.)
| | - Katiuscia Bianchi
- Barts Cancer Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; (V.M.); (K.B.)
| | - Gabor Czibik
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; (L.P.); (G.S.); (M.D.); (C.J.S.); (G.C.)
| | - J. Paul Chapple
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; (L.P.); (G.S.); (M.D.); (C.J.S.); (G.C.)
| |
Collapse
|
17
|
Lv D, Feng P, Guan X, Liu Z, Li D, Xue C, Bai B, Hölscher C. Neuroprotective effects of GLP-1 class drugs in Parkinson's disease. Front Neurol 2024; 15:1462240. [PMID: 39719978 PMCID: PMC11667896 DOI: 10.3389/fneur.2024.1462240] [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: 07/09/2024] [Accepted: 11/14/2024] [Indexed: 12/26/2024] Open
Abstract
Parkinson's disease (PD) is a chronic, progressive neurological disorder primarily affecting motor control, clinically characterized by resting tremor, bradykinesia, rigidity, and other symptoms that significantly diminish the quality of life. Currently, available treatments only alleviate symptoms without halting or delaying disease progression. There is a significant association between PD and type 2 diabetes mellitus (T2DM), possibly due to shared pathological mechanisms such as insulin resistance, chronic inflammation, and mitochondrial dysfunction. PD is caused by a deficiency of dopamine, a neurotransmitter in the brain that plays a critical role in the control of movement. Glucose metabolism and energy metabolism disorders also play an important role in the pathogenesis of PD. This review investigates the neuroprotective mechanisms of glucagon-like peptide-1 (GLP-1) and its receptor agonists, offering novel insights into potential therapeutic strategies for PD. GLP-1 class drugs, primarily used in diabetes management, show promise in addressing PD's underlying pathophysiological mechanisms, including energy metabolism and neuroprotection. These drugs can cross the blood-brain barrier, improve insulin resistance, stabilize mitochondrial function, and enhance neuronal survival and function. Additionally, they exhibit significant anti-inflammatory and antioxidative stress effects, which are crucial in neurodegenerative diseases like PD. Research indicates that GLP-1 receptor agonists could improve both motor and cognitive symptoms in PD patients, marking a potential breakthrough in PD treatment and prevention. Further exploration of GLP-1's molecular mechanisms in PD could provide new preventive and therapeutic approaches, especially for PD patients with concurrent T2DM. By targeting both metabolic and neurodegenerative pathways, GLP-1 receptor agonists represent a multifaceted approach to PD treatment, offering hope for better disease management and improved patient outcomes.
Collapse
Affiliation(s)
- Dongliang Lv
- Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Peng Feng
- Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Xueying Guan
- Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Zhaona Liu
- Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Dongfang Li
- Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Cunshui Xue
- Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Bo Bai
- Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Christian Hölscher
- Henan Academy of Innovations in Medical Science, Brain Institute, Zhengzhou, China
| |
Collapse
|
18
|
Jayakanthan M, Manochkumar J, Efferth T, Ramamoorthy S. Lutein, a versatile carotenoid: Insight on neuroprotective potential and recent advances. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156185. [PMID: 39531935 DOI: 10.1016/j.phymed.2024.156185] [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: 08/27/2024] [Revised: 10/19/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Neurodegenerative diseases (NDDs) are a diverse group of neurological disorders with progressive neuronal loss at specific brain regions, leading to impaired cognitive functioning, loss of neuroplasticity, severe neurological impairment, and dementia. The incidence of neurodegenerative diseases is increasing at an alarming rate with current treatments struggling to barely prolong the inevitable. The desperation to discover a therapeutic agent to treat neurodegenerative diseases and to aid in the process of healthy recovery has opened a gateway into natural pigments. HYPOTHESIS The xanthophyll pigment lutein may bear the potential as a therapeutic agent against NDDs. RESULTS Lutein plays an important role in brain development, cognitive functioning, and improving neuroplasticity. In vitro and in vivo studies revealed the neuroprotective properties of lutein against NDDs such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and cerebral ischemia. The neuroprotective effect of lutein is evidenced by the reduction of free radicals and the simultaneous strengthening of the endogenous antioxidant systems by activating the NRF-2/ERK/AKT pathway. Further, it effectively suppressed mitochondrial aberrations, excitotoxicity, overaccumulation of metals, and its resultant complications. The immunomodulatory activity of lutein prevents neuroinflammation by hindering NF-κB nuclear translocation, regulation of NIK/IKK, PI3K/AKT, MAPK/ERK, JNK pathways, and ICAM-1 downregulation. Lutein also rescued the dysregulated cholinergic system and resolved memory defects. Along with its neuroprotective properties, lutein also improved neuroplasticity by enabling neurogenesis through increased GAP-43, NCAM, and BDNF levels. CONCLUSION Lutein exhibits strong neuroprotective activities against various NDDs. Though the investigations are in the exploratory phase, this review presents the consolidation of scattered evidence of the neuroprotective properties of lutein and urges its further exploration in clinical studies.
Collapse
Affiliation(s)
- Megha Jayakanthan
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Janani Manochkumar
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, 55128, Mainz, Germany
| | - Siva Ramamoorthy
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, India.
| |
Collapse
|
19
|
Abdolizadeh A, Torres-Carmona E, Kambari Y, Amaev A, Song J, Ueno F, Koizumi T, Nakajima S, Agarwal SM, De Luca V, Gerretsen P, Graff-Guerrero A. Evaluation of the Glymphatic System in Schizophrenia Spectrum Disorder Using Proton Magnetic Resonance Spectroscopy Measurement of Brain Macromolecule and Diffusion Tensor Image Analysis Along the Perivascular Space Index. Schizophr Bull 2024; 50:1396-1410. [PMID: 38748498 PMCID: PMC11548937 DOI: 10.1093/schbul/sbae060] [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] [Indexed: 11/10/2024]
Abstract
BACKGROUND AND HYPOTHESIS The glymphatic system (GS), a brain waste clearance pathway, is disrupted in various neurodegenerative and vascular diseases. As schizophrenia shares clinical characteristics with these conditions, we hypothesized GS disruptions in patients with schizophrenia spectrum disorder (SCZ-SD), reflected in increased brain macromolecule (MM) and decreased diffusion-tensor-image-analysis along the perivascular space (DTI-ALPS) index. STUDY DESIGN Forty-seven healthy controls (HCs) and 103 patients with SCZ-SD were studied. Data included 135 proton magnetic resonance spectroscopy (1H-MRS) sets, 96 DTI sets, with 79 participants contributing both. MM levels were quantified in the dorsal-anterior cingulate cortex (dACC), dorsolateral prefrontal cortex, and dorsal caudate (point resolved spectroscopy, echo-time = 35ms). Diffusivities in the projection and association fibers near the lateral ventricle were measured to calculate DTI-ALPS indices. General linear models were performed, adjusting for age, sex, and smoking. Correlation analyses examined relationships with age, illness duration, and symptoms severity. STUDY RESULTS MM levels were not different between patients and HCs. However, left, right, and bilateral DTI-ALPS indices were lower in patients compared with HCs (P < .001). In HCs, age was positively correlated with dACC MM and negatively correlated with left, right, and bilateral DTI-ALPS indices (P < .001). In patients, illness duration was positively correlated with dACC MM and negatively correlated with the right DTI-ALPS index (P < .05). In the entire population, dACC MM and DTI-ALPS indices showed an inverse correlation (P < .01). CONCLUSIONS Our results suggest potential disruptions in the GS of patients with SCZ-SD. Improving brain's waste clearance may offer a potential therapeutic approach for patients with SCZ-SD.
Collapse
Affiliation(s)
- Ali Abdolizadeh
- Multimodal Imaging Group, Research Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Edgardo Torres-Carmona
- Multimodal Imaging Group, Research Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Yasaman Kambari
- Multimodal Imaging Group, Research Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Aron Amaev
- Multimodal Imaging Group, Research Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jianmeng Song
- Multimodal Imaging Group, Research Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Fumihiko Ueno
- Multimodal Imaging Group, Research Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Teruki Koizumi
- Multimodal Imaging Group, Research Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, National Hospital Organization Shimofusa Psychiatric Medical Center, Chiba, Japan
| | - Shinichiro Nakajima
- Multimodal Imaging Group, Research Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Sri Mahavir Agarwal
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Vincenzo De Luca
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Philip Gerretsen
- Multimodal Imaging Group, Research Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, CAMH, Toronto, ON, Canada
| | - Ariel Graff-Guerrero
- Multimodal Imaging Group, Research Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, CAMH, Toronto, ON, Canada
| |
Collapse
|
20
|
Ye Y, Fu C, Li Y, Sun J, Li X, Chai S, Li S, Hou M, Cai H, Wang Z, Wu M. Alternate-day fasting improves cognitive and brain energy deficits by promoting ketone metabolism in the 3xTg mouse model of Alzheimer's disease. Exp Neurol 2024; 381:114920. [PMID: 39142368 DOI: 10.1016/j.expneurol.2024.114920] [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: 03/21/2024] [Revised: 07/10/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024]
Abstract
Alzheimer's disease (AD) is characterized by disorders in brain energy. The lack of sufficient energy for nerve function leads to cognitive dysfunction and massive neuronal loss in AD. Ketone bodies are an alternative to glucose as a source of energy in the brain, and alternate-day fasting (ADF) promotes the production of the ketone body β-hydroxybutyric acid (βOHB). In this study, 7-month-old male WT mice and 3xTg mice underwent dietary control for 20 weeks. We found that ADF increased circulating βOHB concentrations in 3xTg mice, improved cognitive function, reduced anxiety-like behaviors, improved hippocampal synaptic plasticity, and reduced neuronal loss, Aβ oligomers and tau hyperphosphorylation. In addition, ADF improved mitochondrial bioenergetic function by promoting brain ketone metabolism and rescued brain energy deficits in 3xTg mice. A safety evaluation showed that ADF improved exercise endurance and liver and kidney function in 3xTg mice without negatively affecting muscle motor and heart functions. This study provides a theoretical basis and strong support for the application of ADF as a non-drug strategy for preventing and treating brain energy defects in the early stage of AD.
Collapse
Affiliation(s)
- Yucai Ye
- Department of Physiology, School of Basic Medicine, Shanxi Medical University, Key Laboratory of Cellular Physiology, Ministry of Education, Taiyuan 030001, China
| | - Chaojing Fu
- Department of Physiology, School of Basic Medicine, Shanxi Medical University, Key Laboratory of Cellular Physiology, Ministry of Education, Taiyuan 030001, China
| | - Yan Li
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan 030001, China; Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Junli Sun
- School of Anesthesiology, Shanxi Medical University, Taiyuan 030001, China
| | - Xinru Li
- Department of Physiology, School of Basic Medicine, Shanxi Medical University, Key Laboratory of Cellular Physiology, Ministry of Education, Taiyuan 030001, China
| | - Shifan Chai
- Department of Physiology, School of Basic Medicine, Shanxi Medical University, Key Laboratory of Cellular Physiology, Ministry of Education, Taiyuan 030001, China
| | - Shuo Li
- Second Clinical Medical College, Shanxi Medical University, Taiyuan 030001, China
| | - Meng Hou
- Second Clinical Medical College, Shanxi Medical University, Taiyuan 030001, China
| | - Hongyan Cai
- Department of Microbiology and Immunology, School of Basic Medicine, Shanxi Medical University, Taiyuan 030001, China
| | - Zhaojun Wang
- Department of Physiology, School of Basic Medicine, Shanxi Medical University, Key Laboratory of Cellular Physiology, Ministry of Education, Taiyuan 030001, China.
| | - Meina Wu
- Department of Physiology, School of Basic Medicine, Shanxi Medical University, Key Laboratory of Cellular Physiology, Ministry of Education, Taiyuan 030001, China.
| |
Collapse
|
21
|
Ramires Júnior OV, Silveira JS, Gusso D, Krupp Prauchner GR, Ferrary Deniz B, Almeida WD, Pereira LO, Wyse AT. Homocysteine decreases VEGF, EGF, and TrkB levels and increases CCL5/RANTES in the hippocampus: Neuroprotective effects of rivastigmine and ibuprofen. Chem Biol Interact 2024; 403:111260. [PMID: 39357784 DOI: 10.1016/j.cbi.2024.111260] [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: 04/30/2024] [Revised: 09/21/2024] [Accepted: 09/30/2024] [Indexed: 10/04/2024]
Abstract
Homocysteine (Hcy) is produced through methionine transmethylation. Elevated Hcy levels are termed Hyperhomocysteinemia (HHcy) and represent a risk factor for neurodegenerative conditions such as Alzheimer's disease. This study aimed to explore the impact of mild HHcy and the neuroprotective effects of ibuprofen and rivastigmine via immunohistochemical analysis of glial markers (Iba-1 and GFAP). Additionally, we assessed levels of vascular endothelial growth factor (VEGF), epidermal growth factor (EGF), chemokine ligand 5 (CCL5/RANTES), CX3C chemokine ligand 1 (CX3CL1), and the NGF/p75NTR/tropomyosin kinase B (TrkB) pathway in the hippocampus of adult rats. Mild chronic HHcy was induced chemically in Wistar rats by subcutaneous administration of Hcy (4 mg/kg body weight) twice daily for 30 days. Rivastigmine (0.5 mg/kg) and ibuprofen (40 mg/kg) were administered intraperitoneally once daily. Results revealed elevated levels of CCL5/RANTES and reduced levels of VEGF, EGF, and TrkB in the hippocampus of HHcy-exposed rats. Rivastigmine mitigated the neurotoxic effects of HHcy by increasing TrkB and VEGF levels. Conversely, ibuprofen attenuated CCL5/RANTES levels against the neurotoxicity of HHcy, significantly reducing this chemokine's levels. HHcy-induced neurochemical impairment in the hippocampus may jeopardize neurogenesis, synapse formation, axonal transport, and inflammatory balance, leading to neurodegeneration. Treatments with rivastigmine and ibuprofen alleviated some of these detrimental effects. Reversing HHcy-induced damage through these compounds could serve as a potential neuroprotective strategy against brain damage.
Collapse
Affiliation(s)
- Osmar Vieira Ramires Júnior
- Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratory of Neuroprotection and Neurometabolic Diseases, Department of Biochemistry, ICBS, Federal University of Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Zip code 90035003, Porto Alegre, RS, Brazil
| | - Josiane Silva Silveira
- Laboratory of Neuroprotection and Neurometabolic Diseases, Department of Biochemistry, ICBS, Federal University of Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Zip code 90035003, Porto Alegre, RS, Brazil
| | - Darlan Gusso
- Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratory of Neuroprotection and Neurometabolic Diseases, Department of Biochemistry, ICBS, Federal University of Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Zip code 90035003, Porto Alegre, RS, Brazil
| | - Gustavo Ricardo Krupp Prauchner
- Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratory of Neuroprotection and Neurometabolic Diseases, Department of Biochemistry, ICBS, Federal University of Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Zip code 90035003, Porto Alegre, RS, Brazil
| | - Bruna Ferrary Deniz
- Departamento de Ciências Morfológicas, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Departamento de Fisiologia e Farmacologia, Instiruto de Biologia, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Wellington de Almeida
- Program in Neurosciences, ICBS, Federal Universityof Rio Grande do Sul, Porto Alegre, RS, Brazil; Departamento de Ciências Morfológicas, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Lenir Orlandi Pereira
- Program in Neurosciences, ICBS, Federal Universityof Rio Grande do Sul, Porto Alegre, RS, Brazil; Departamento de Ciências Morfológicas, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Angela Ts Wyse
- Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratory of Neuroprotection and Neurometabolic Diseases, Department of Biochemistry, ICBS, Federal University of Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Zip code 90035003, Porto Alegre, RS, Brazil.
| |
Collapse
|
22
|
Feng Y, Hao F. Advances in natural polysaccharides in Alzheimer's disease and Parkinson's disease: Insights from the brain-gut axis. Trends Food Sci Technol 2024; 153:104678. [DOI: 10.1016/j.tifs.2024.104678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
23
|
Aslim B, Nigdelioglu Dolanbay S, Baran SS. Exploring allocryptopine as a neuroprotective agent against oxidative stress-induced neural apoptosis via Akt/GSK-3β/tau pathway modulation. Comput Biol Chem 2024; 112:108144. [PMID: 39004026 DOI: 10.1016/j.compbiolchem.2024.108144] [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: 02/20/2024] [Revised: 06/20/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024]
Abstract
Alzheimer's disease (AD) is characterized by neuronal loss due to hyperphosphorylated proteins induced by oxidative stress. AD remains a formidable challenge in the medical field, as current treatments focusing on single biomarkers have yielded limited success. Hence, there's a burgeoning interest in investigating novel compounds that can target mechanisms, offering alternative therapeutic approaches. The aim of this study is to investigate the effects of allocryptopine, an isoquinoline alkaloid, on mechanisms related to AD in order to develop alternative treatment strategies. In this study, the in vitro AD cell model was obtained by inducing nerve growth factor (NGF)-differentiated PC12 (dPC12) cells to oxidative stress with H2O2, and also the effect mechanism of different allocryptopine concentrations on the in vitro AD cell model was studied. The treatments' antioxidative effects at the ROS level and their regulation of the cell cycle were assessed through flow cytometry, while their anti-apoptotic effects were evaluated using both flow cytometry and qRT-PCR. Additionally, the phosphorylation levels of Akt, GSK-3β, and tau proteins were analyzed via western blot, and the interactions between Akt, GSK-3β, CDK5 proteins, and allocryptopine were demonstrated through molecular docking. Our study's conclusive results revealed that allocryptopine effectively suppressed intracellular ROS levels, while simultaneously enhancing the Akt/GSK-3β signaling pathway by increasing p-Akt and p-GSK-3β proteins. This mechanism played a critical role in inhibiting neural cell apoptosis and preventing tau hyperphosphorylation. Moreover, allocryptopine demonstrated its ability to regulate the G1/S cell cycle progression, leading to cell cycle arrest in the G1 phase, and facilitating cellular repair mechanisms, potentially contributing to the suppression of neural apoptosis. The in silico results of allocryptopine were shown to docking with the cyclin-dependent kinase 5 (CDK 5) playing a role in tau phosphorylation Akt and GSK-3β from target proteins. Therefore, the in silico study results supported the in vitro results. The results showed that allocryptopine can protect dPC12 cells from oxidative stress-induced apoptosis and hyperphosphorylation of the tau protein by regulating the Akt/GSK-3β signaling pathway. Based on these findings, it can be suggested that allocryptopine, with its ability to target biomarkers and its significant effects on AD-associated mechanisms, holds promise as a potential candidate for drug development in the treatment of AD. Further research and clinical trials are recommended in the future.
Collapse
Affiliation(s)
- Belma Aslim
- Gazi University, Faculty of Science, Department of Biology, Ankara 06500, Turkey
| | | | - Sahra Setenay Baran
- Gazi University, Faculty of Science, Department of Biology, Ankara 06500, Turkey; Gazi University, Graduate School of Natural and Applied Sciences, Department of Biology, Ankara 06500, Turkey
| |
Collapse
|
24
|
Ranjan A, Gandhi SR. Propagation of transient explosive synchronization in a mesoscale mouse brain network model of epilepsy. Netw Neurosci 2024; 8:883-901. [PMID: 39355439 PMCID: PMC11398721 DOI: 10.1162/netn_a_00379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 04/18/2024] [Indexed: 10/03/2024] Open
Abstract
Generalized epileptic attacks, which exhibit widespread disruption of brain activity, are characterized by recurrent, spontaneous, and synchronized bursts of neural activity that self-initiate and self-terminate through critical transitions. Here we utilize the general framework of explosive synchronization (ES) from complex systems science to study the role of network structure and resource dynamics in the generation and propagation of seizures. We show that a combination of resource constraint and adaptive coupling in a Kuramoto network oscillator model can reliably generate seizure-like synchronization activity across different network topologies, including a biologically derived mesoscale mouse brain network. The model, coupled with a novel algorithm for tracking seizure propagation, provides mechanistic insight into the dynamics of transition to the synchronized state and its dependence on resources; and identifies key brain areas that may be involved in the initiation and spatial propagation of the seizure. The model, though minimal, efficiently recapitulates several experimental and theoretical predictions from more complex models and makes novel experimentally testable predictions.
Collapse
Affiliation(s)
- Avinash Ranjan
- Department of Electrical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Saurabh R Gandhi
- Department of Electrical Engineering, Indian Institute of Technology Delhi, New Delhi, India
- Center for Brain Science and Applications, School of Artificial Intelligence and Data Science, Indian Institute of Technology, Jodhpur, India
| |
Collapse
|
25
|
Rodriguez P, Kalia V, Fenollar-Ferrer C, Gibson CL, Gichi Z, Rajoo A, Matier CD, Pezacki AT, Xiao T, Carvelli L, Chang CJ, Miller GW, Khamoui AV, Boerner J, Blakely RD. Glial swip-10 controls systemic mitochondrial function, oxidative stress, and neuronal viability via copper ion homeostasis. Proc Natl Acad Sci U S A 2024; 121:e2320611121. [PMID: 39288174 PMCID: PMC11441482 DOI: 10.1073/pnas.2320611121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 08/01/2024] [Indexed: 09/19/2024] Open
Abstract
Cuprous copper [Cu(I)] is an essential cofactor for enzymes that support many fundamental cellular functions including mitochondrial respiration and suppression of oxidative stress. Neurons are particularly reliant on mitochondrial production of ATP, with many neurodegenerative diseases, including Parkinson's disease, associated with diminished mitochondrial function. The gene MBLAC1 encodes a ribonuclease that targets pre-mRNA of replication-dependent histones, proteins recently found in yeast to reduce Cu(II) to Cu(I), and when mutated disrupt ATP production, elevates oxidative stress, and severely impacts cell growth. Whether this process supports neuronal and/or systemic physiology in higher eukaryotes is unknown. Previously, we identified swip-10, the putative Caenorhabditis elegans ortholog of MBLAC1, establishing a role for glial swip-10 in limiting dopamine (DA) neuron excitability and sustaining DA neuron viability. Here, we provide evidence from computational modeling that SWIP-10 protein structure mirrors that of MBLAC1 and locates a loss of function coding mutation at a site expected to disrupt histone RNA hydrolysis. Moreover, we find through genetic, biochemical, and pharmacological studies that deletion of swip-10 in worms negatively impacts systemic Cu(I) levels, leading to deficits in mitochondrial respiration and ATP production, increased oxidative stress, and neurodegeneration. These phenotypes can be offset in swip-10 mutants by the Cu(I) enhancing molecule elesclomol and through glial expression of wildtype swip-10. Together, these studies reveal a glial-expressed pathway that supports systemic mitochondrial function and neuronal health via regulation of Cu(I) homeostasis, a mechanism that may lend itself to therapeutic strategies to treat devastating neurodegenerative diseases.
Collapse
Affiliation(s)
- Peter Rodriguez
- Department of Biological Sciences, Charles E. Schmidt College of Science, Boca Raton, FL33412
| | - Vrinda Kalia
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY10032
| | - Cristina Fenollar-Ferrer
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, Bethesda, MD20892
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL33458
| | - Chelsea L. Gibson
- Department of Biological Sciences, Charles E. Schmidt College of Science, Boca Raton, FL33412
- Oak Ridge Institute for Science and Education, Oak Ridge, TN37830
| | - Zayna Gichi
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Jupiter, FL33458
| | - Andre Rajoo
- Department of Biology, Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, FL33458
| | - Carson D. Matier
- Department of Chemistry, University of California, Berkeley, CA94720
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
| | - Aidan T. Pezacki
- Department of Chemistry, University of California, Berkeley, CA94720
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
- Princeton University, Princeton, NJ08544
| | - Tong Xiao
- Department of Chemistry, University of California, Berkeley, CA94720
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
- Princeton University, Princeton, NJ08544
| | - Lucia Carvelli
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL33458
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Jupiter, FL33458
- Department of Biology, Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, FL33458
| | - Christopher J. Chang
- Department of Chemistry, University of California, Berkeley, CA94720
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
- Princeton University, Princeton, NJ08544
| | - Gary W. Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY10032
| | - Andy V. Khamoui
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL33458
- Department of Exercise Science and Health Promotion, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, FL33431
| | - Jana Boerner
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL33458
| | - Randy D. Blakely
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL33458
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Jupiter, FL33458
| |
Collapse
|
26
|
Allboani A, Kar S, Kavdia M. Computational modeling of neuronal nitric oxide synthase biochemical pathway: A mechanistic analysis of tetrahydrobiopterin and oxidative stress. Free Radic Biol Med 2024; 222:625-637. [PMID: 39004235 DOI: 10.1016/j.freeradbiomed.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/14/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
Neuronal cell dysfunction plays an important role in neurodegenerative diseases. Oxidative stress can disrupt the redox balance within neuronal cells and may cause neuronal nitric oxide synthase (nNOS) to uncouple, contributing to the neurodegenerative processes. Experimental studies and clinical trials using nNOS cofactor tetrahydrobiopterin (BH4) and antioxidants in neuronal cell dysfunction have shown inconsistent results. A better mechanistic understanding of complex interactions of nNOS activity and oxidative stress in neuronal cell dysfunction is needed. In this study, we developed a computational model of neuronal cell using nNOS biochemical pathways to explore several key mechanisms that are known to influence neuronal cell redox homeostasis. We studied the effects of oxidative stress and BH4 synthesis on nNOS nitric oxide production and biopterin ratio (BH4/total biopterin). Results showed that nNOS remained coupled and maintained nitric oxide production for oxidative stress levels less than 230 nM/s. The results showed that neuronal oxidative stress above 230 nM/s increased the degree of nNOS uncoupling and introduced instability in the nitric oxide production. The nitric oxide production did not change irrespective of initial biopterin ratio of 0.05-0.99 for a given oxidative stress. Oxidative stress resulted in significant reduction in BH4 levels even when nitric oxide production was not affected. Enhancing BH4 synthesis or supplementation improved nNOS coupling, however the degree of improvement was determined by the levels of oxidative stress and BH4 synthesis. The results of our mechanistic analysis indicate that there is a potential for significant improvement in neuronal dysfunction by simultaneously increasing BH4 levels and reducing cellular oxidative stress.
Collapse
Affiliation(s)
- Amnah Allboani
- Department of Biomedical Engineering, Wayne State University, Detroit, MI, 48202, USA
| | - Saptarshi Kar
- College of Engineering and Technology, American University of the Middle East, Kuwait
| | - Mahendra Kavdia
- Department of Biomedical Engineering, Wayne State University, Detroit, MI, 48202, USA.
| |
Collapse
|
27
|
Wang S, Chen Y, Xiong L, Jin N, Zhao P, Liang Z, Cheng L, Kang L. Multimorbidity measures associated with cognitive function among community-dwelling older Chinese adults. Alzheimers Dement 2024; 20:6221-6231. [PMID: 39072982 PMCID: PMC11497665 DOI: 10.1002/alz.14117] [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: 07/11/2023] [Revised: 05/17/2024] [Accepted: 06/07/2024] [Indexed: 07/30/2024]
Abstract
INTRODUCTION Older adults with multimorbidity are at high risk of cognitive impairment development. There is a lack of research on the associations between different multimorbidity measures and cognitive function among older Chinese adults living in the community. METHODS We used the Chinese Longitudinal Healthy Longevity Survey from 2002 to 2018 and included data on dementia-free participants aged ≥65 years. Multimorbidity measures included condition counts, multimorbidity patterns, and trajectories. The association of multimorbidity measures with cognitive function was examined by generalized estimating equation and linear and logistic regression models. RESULTS Among 14,093 participants at baseline, 43.2% had multimorbidity. Multimorbidity patterns were grouped into cancer-inflammatory, cardiometabolic, and sensory patterns. Multimorbidity trajectories were classified as "onset-condition," "newly developing," and "severe condition." The Mini-Mental State Examination scores were significantly lower for participants with more chronic conditions, with cancer-inflammatory/cardiometabolic/sensory patterns, and with developing multimorbidity trajectories. DISCUSSION Condition counts, sensory pattern, cardiometabolic pattern, cancer-inflammatory pattern, and multimorbidity developmental trajectories were prospectively associated with cognitive function. HIGHLIGHTS Elderly individuals with a higher number of chronic conditions were associated with lower MMSE scores in the Chinese Longitudinal Healthy Longevity Survey data. MMSE scores were significantly lower for participants with specific multimorbidity patterns. Individuals with developing trajectories of multimorbidity were associated with lower MMSE scores and a higher risk of mild cognitive impairment.
Collapse
Affiliation(s)
- Shuojia Wang
- Department of GeriatricsGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Center for GeriatricsShenzhen People's Hospital (The Second Clinical Medical College, Jinan University)ShenzhenChina
- Post‐doctoral Scientific Research Station of Basic MedicineJinan UniversityGuangzhouChina
| | - Yilin Chen
- Department of GeriatricsGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Center for GeriatricsShenzhen People's Hospital (The Second Clinical Medical College, Jinan University)ShenzhenChina
| | - Lijiao Xiong
- Department of GeriatricsGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Center for GeriatricsShenzhen People's Hospital (The Second Clinical Medical College, Jinan University)ShenzhenChina
| | - Nana Jin
- Department of GeriatricsGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Center for GeriatricsShenzhen People's Hospital (The Second Clinical Medical College, Jinan University)ShenzhenChina
- Post‐doctoral Scientific Research Station of Basic MedicineJinan UniversityGuangzhouChina
| | - Pengfei Zhao
- Department of GeriatricsGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Center for GeriatricsShenzhen People's Hospital (The Second Clinical Medical College, Jinan University)ShenzhenChina
| | - Zhen Liang
- Department of GeriatricsGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Center for GeriatricsShenzhen People's Hospital (The Second Clinical Medical College, Jinan University)ShenzhenChina
| | - Lixin Cheng
- Department of GeriatricsGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Center for GeriatricsShenzhen People's Hospital (The Second Clinical Medical College, Jinan University)ShenzhenChina
| | - Lin Kang
- Department of GeriatricsGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Center for GeriatricsShenzhen People's Hospital (The Second Clinical Medical College, Jinan University)ShenzhenChina
| |
Collapse
|
28
|
Georg Jensen M, Goode M, Heinrich M. Herbal medicines and botanicals for managing insomnia, stress, anxiety, and depression: A critical review of the emerging evidence focusing on the Middle East and Africa. PHARMANUTRITION 2024; 29:100399. [DOI: 10.1016/j.phanu.2024.100399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
29
|
Zheng T, Kotol D, Sjöberg R, Mitsios N, Uhlén M, Zhong W, Edfors F, Mulder J. Characterization of reduced astrocyte creatine kinase levels in Alzheimer's disease. Glia 2024; 72:1590-1603. [PMID: 38856187 DOI: 10.1002/glia.24569] [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: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 06/11/2024]
Abstract
The creatine-phosphocreatine cycle serves as a crucial temporary energy buffering system in the brain, regulated by brain creatine kinase (CKB), in maintaining Adenosine triphosphate (ATP) levels. Alzheimer's disease (AD) has been linked to increased CKB oxidation and loss of its regulatory function, although specific pathological processes and affected cell types remain unclear. In our study, cerebral cortex samples from individuals with AD, dementia with Lewy bodies (DLB), and age-matched controls were analyzed using antibody-based methods to quantify CKB levels and assess alterations associated with disease processes. Two independently validated antibodies exclusively labeled astrocytes in the human cerebral cortex. Combining immunofluorescence (IF) and mass spectrometry (MS), we explored CKB availability in AD and DLB cases. IF and Western blot analysis demonstrated a loss of CKB immunoreactivity correlated with increased plaque load, severity of tau pathology, and Lewy body pathology. However, transcriptomics data and targeted MS demonstrated unaltered total CKB levels, suggesting posttranslational modifications (PTMs) affecting antibody binding. This aligns with altered efficiency at proteolytic cleavage sites indicated in the targeted MS experiment. These findings highlight that the proper function of astrocytes, understudied in the brain compared with neurons, is highly affected by PTMs. Reduction in ATP levels within astrocytes can disrupt ATP-dependent processes, such as the glutamate-glutamine cycle. As CKB and the creatine-phosphocreatine cycle are important in securing constant ATP availability, PTMs in CKB, and astrocyte dysfunction may disturb homeostasis, driving excitotoxicity in the AD brain. CKB and its activity could be promising biomarkers for monitoring early-stage energy deficits in AD.
Collapse
Affiliation(s)
- Tianyu Zheng
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - David Kotol
- Department of Proteomics and Nanobiotechnology, Royal Institute of Technology, Stockholm, Sweden
| | - Rebecca Sjöberg
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Nicholas Mitsios
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Mathias Uhlén
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Proteomics and Nanobiotechnology, Royal Institute of Technology, Stockholm, Sweden
| | - Wen Zhong
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Science for Life Laboratory, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Fredrik Edfors
- Department of Proteomics and Nanobiotechnology, Royal Institute of Technology, Stockholm, Sweden
| | - Jan Mulder
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
30
|
Sharika R, Mongkolpobsin K, Rangsinth P, Prasanth MI, Nilkhet S, Pradniwat P, Tencomnao T, Chuchawankul S. Experimental Models in Unraveling the Biological Mechanisms of Mushroom-Derived Bioactives against Aging- and Lifestyle-Related Diseases: A Review. Nutrients 2024; 16:2682. [PMID: 39203820 PMCID: PMC11357205 DOI: 10.3390/nu16162682] [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: 07/14/2024] [Revised: 07/29/2024] [Accepted: 08/10/2024] [Indexed: 09/03/2024] Open
Abstract
Mushrooms have garnered considerable interest among researchers due to their immense nutritional and therapeutic properties. The presence of biologically active primary and secondary metabolites, which includes several micronutrients, including vitamins, essential minerals, and other dietary fibers, makes them an excellent functional food. Moreover, the dietary inclusion of mushrooms has been reported to reduce the incidence of aging- and lifestyle-related diseases, such as cancer, obesity, and stroke, as well as to provide overall health benefits by promoting immunomodulation, antioxidant activity, and enhancement of gut microbial flora. The multifunctional activities of several mushroom extracts have been evaluated by both in vitro and in vivo studies using cell lines along with invertebrate and vertebrate model systems to address human diseases and disorders at functional and molecular levels. Although each model has its own strengths as well as lacunas, various studies have generated a plethora of data regarding the regulating players that are modulated in order to provide various protective activities; hence, this review intends to compile and provide an overview of the plausible mechanism of action of mushroom-derived bioactives, which will be helpful in future medicinal explorations.
Collapse
Affiliation(s)
- Rajasekharan Sharika
- Immunomodulation of Natural Products Research Unit, Chulalongkorn University, Bangkok 10330, Thailand; (R.S.); (K.M.); (S.N.); (P.P.)
- Department of Transfusion Medicine and Clinical Microbiology, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Kuljira Mongkolpobsin
- Immunomodulation of Natural Products Research Unit, Chulalongkorn University, Bangkok 10330, Thailand; (R.S.); (K.M.); (S.N.); (P.P.)
- Department of Transfusion Medicine and Clinical Microbiology, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Panthakarn Rangsinth
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China;
| | - Mani Iyer Prasanth
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok 10330, Thailand; (M.I.P.); (T.T.)
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Sunita Nilkhet
- Immunomodulation of Natural Products Research Unit, Chulalongkorn University, Bangkok 10330, Thailand; (R.S.); (K.M.); (S.N.); (P.P.)
- Department of Transfusion Medicine and Clinical Microbiology, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Paweena Pradniwat
- Immunomodulation of Natural Products Research Unit, Chulalongkorn University, Bangkok 10330, Thailand; (R.S.); (K.M.); (S.N.); (P.P.)
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok 10330, Thailand; (M.I.P.); (T.T.)
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Siriporn Chuchawankul
- Immunomodulation of Natural Products Research Unit, Chulalongkorn University, Bangkok 10330, Thailand; (R.S.); (K.M.); (S.N.); (P.P.)
- Department of Transfusion Medicine and Clinical Microbiology, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
31
|
Nair SS, Chakravarthy S. A Computational Model of Deep Brain Stimulation for Parkinson's Disease Tremor and Bradykinesia. Brain Sci 2024; 14:620. [PMID: 38928620 PMCID: PMC11201485 DOI: 10.3390/brainsci14060620] [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: 04/29/2024] [Revised: 06/05/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Parkinson's disease (PD) is a progressive neurological disorder that is typically characterized by a range of motor dysfunctions, and its impact extends beyond physical abnormalities into emotional well-being and cognitive symptoms. The loss of dopaminergic neurons in the substantia nigra pars compacta (SNc) leads to an array of dysfunctions in the functioning of the basal ganglia (BG) circuitry that manifests into PD. While active research is being carried out to find the root cause of SNc cell death, various therapeutic techniques are used to manage the symptoms of PD. The most common approach in managing the symptoms is replenishing the lost dopamine in the form of taking dopaminergic medications such as levodopa, despite its long-term complications. Another commonly used intervention for PD is deep brain stimulation (DBS). DBS is most commonly used when levodopa medication efficacy is reduced, and, in combination with levodopa medication, it helps reduce the required dosage of medication, prolonging the therapeutic effect. DBS is also a first choice option when motor complications such as dyskinesia emerge as a side effect of medication. Several studies have also reported that though DBS is found to be effective in suppressing severe motor symptoms such as tremors and rigidity, it has an adverse effect on cognitive capabilities. Henceforth, it is important to understand the exact mechanism of DBS in alleviating motor symptoms. A computational model of DBS stimulation for motor symptoms will offer great insights into understanding the mechanisms underlying DBS, and, along this line, in our current study, we modeled a cortico-basal ganglia circuitry of arm reaching, where we simulated healthy control (HC) and PD symptoms as well as the DBS effect on PD tremor and bradykinesia. Our modeling results reveal that PD tremors are more correlated with the theta band, while bradykinesia is more correlated with the beta band of the frequency spectrum of the local field potential (LFP) of the subthalamic nucleus (STN) neurons. With a DBS current of 220 pA, 130 Hz, and a 100 microsecond pulse-width, we could found the maximum therapeutic effect for the pathological dynamics simulated using our model using a set of parameter values. However, the exact DBS characteristics vary from patient to patient, and this can be further studied by exploring the model parameter space. This model can be extended to study different DBS targets and accommodate cognitive dynamics in the future to study the impact of DBS on cognitive symptoms and thereby optimize the parameters to produce optimal performance effects across modalities. Combining DBS with rehabilitation is another frontier where DBS can reduce symptoms such as tremors and rigidity, enabling patients to participate in their therapy. With DBS providing instant relief to patients, a combination of DBS and rehabilitation can enhance neural plasticity. One of the key motivations behind combining DBS with rehabilitation is to expect comparable results in motor performance even with milder DBS currents.
Collapse
Affiliation(s)
| | - Srinivasa Chakravarthy
- Department of Biotechnology, Bhupat and Mehta Jyoti School of Biosciences, Chennai 600036, India;
- Department of Medical Science and Technology, Indian Institute of Technology Madras, Sardar Patel Road, Adyar, Chennai 600036, India
| |
Collapse
|
32
|
Oliai SF, Shippy DC, Ulland TK. Mitigation of CXCL10 secretion by metabolic disorder drugs in microglial-mediated neuroinflammation. J Neuroimmunol 2024; 391:578364. [PMID: 38718558 PMCID: PMC11165694 DOI: 10.1016/j.jneuroim.2024.578364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/26/2024] [Accepted: 05/02/2024] [Indexed: 05/12/2024]
Abstract
Metabolic disorders are associated with several neurodegenerative diseases. We previously identified C-X-C motif chemokine ligand 10 (CXCL10), also known as interferon gamma-induced protein 10 (IP-10), as a major contributor to the type I interferon response in microglial-mediated neuroinflammation. Therefore, we hypothesized FDA-approved metabolic disorder drugs that attenuate CXCL10 secretion may be repurposed as a treatment for neurodegenerative diseases. Screening, dose curves, and cytotoxicity assays in LPS-stimulated microglia yielded treprostinil (hypertension), pitavastatin (hyperlipidemia), and eplerenone (hypertension) as candidates that significantly reduced CXCL10 secretion (in addition to other pro-inflammatory mediators) without impacting cell viability. Altogether, these data suggest metabolic disorder drugs that attenuate CXCL10 as potential treatments for neurodegenerative disease through mitigating microglial-mediated neuroinflammation.
Collapse
Affiliation(s)
- Sophia F Oliai
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Daniel C Shippy
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Tyler K Ulland
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
33
|
Ahmed A, Khan AU, Nadeem H, Imran M, Irshad N. Pharmacological evaluation of newly synthesized benzimidazole derivative for anti-Alzheimer potential. Int J Neurosci 2024; 134:635-651. [PMID: 36259511 DOI: 10.1080/00207454.2022.2138382] [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: 07/13/2022] [Revised: 09/16/2022] [Accepted: 09/26/2022] [Indexed: 01/01/2023]
Abstract
Backgound: Alzheimer disease (AD) is a disastrous disease characterized by accretion of amyloid-beta plaques, neurofibrillary tangles inducing oxidative stress, loss of neuronal functions and continuous progression of cognitive impairment leading to severe dementia. Material and Methods: The newly synthesized benzimidazole derivative 4-chloro-3-(2-phenyl-1H-benzimidazole-1-sulfonyl) benzoic acid (CB) was evaluated for its anti-Alzheimer activity using in silico, in vivo, in vitro and molecular techniques (ELISA, WB & IHC). Results: In-silico studies revealed that CB has atomic contact energy values of -3.9 to -8.9 kcal/mol against selected targets. In vitro assay showed that CB caused acetylcholinesterase (AChE) and diphenyl-1-picrylhydrazyl inhibition. In-vivo findings revealed improvement in dementia as observed in the morris water maze test and Ymaze test. Amyloid-beta disaggregation, increased level of anti-oxidants, decreased expressions of inflammatory markers and enhanced cellular architecture were found in the cortex and hippocampus of treated rats in the histopathological examination, immunohistochemistry analysis, enzyme-linked immunosorbent assay and western blot analysis. Conclusions: This study revealed that CB possess different binding affinities with the Alzheimer-related targets and it possess anti-Alzheimer activity, mediated via AChE and amyloid-beta inhibition, anti-oxidant and anti-inflammatory pathways.
Collapse
Affiliation(s)
- Aleeza Ahmed
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Arif-Ullah Khan
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Humaira Nadeem
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Muhammad Imran
- Iqra Department Of Pharmacy, Iqra University, Islamabad, Pakistan
| | - Nadeem Irshad
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, Pakistan
| |
Collapse
|
34
|
Zaib S, Khan I, Ali HS, Younas MT, Ibrar A, Al-Odayni AB, Al-Kahtani AA. Design and discovery of anthranilamide derivatives as a potential treatment for neurodegenerative disorders via targeting cholinesterases and monoamine oxidases. Int J Biol Macromol 2024; 272:132748. [PMID: 38821306 DOI: 10.1016/j.ijbiomac.2024.132748] [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: 03/04/2024] [Revised: 05/26/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Neurodegenerative diseases with progressive cellular loss of the central nervous system and elusive disease etiology provide a continuous impetus to explore drug discovery programmes aiming at identifying robust and effective inhibitors of cholinesterase and monoamine oxidase enzymes. We herein present a concise library of anthranilamide derivatives involving a palladium-catalyzed Suzuki-Miyaura cross-coupling reaction to install the diverse structural diversity required for the desired biological action. Using Ellman's method, cholinesterase inhibitory activity was performed against AChE and BuChE enzymes. In vitro assay results demonstrated that anthranilamides are potent inhibitors with remarkable potency. Compound 6k emerged as the lead candidate and dual inhibitor of both enzymes with IC50 values of 0.12 ± 0.01 and 0.49 ± 0.02 μM against AChE and BuChE, respectively. Several other compounds were found as highly potent and selective inhibitors. Anthranilamide derivatives were also tested against monoamine oxidase (A and B) enzymes using fluorometric method. In vitro data revealed compound 6h as the most potent inhibitor against MAO-A, showing an IC50 value of 0.44 ± 0.02 μM, whereas, compound 6k emerged as the top inhibitor of MAO-B with an IC50 value of 0.06 ± 0.01 μM. All the lead inhibitors were analyzed for the identification of their mechanism of action using Michaelis-Menten kinetics experiments. Compound 6k and 6h depicted a competitive mode of action against AChE and MAO-A, whereas, a non-competitive and mixed-type of inhibition was observed against BuChE and MAO-B by compounds 6k. Molecular docking analysis revealed remarkable binding affinities of the potent inhibitors with specific residues inside the active site of receptors. Furthermore, molecular dynamics simulations were performed to explore the ability of potent compounds to form energetically stable complexes with the target protein. Finally, in silico ADME calculations also demonstrated that the potent compounds exhibit promising pharmacokinetic profile, satisfying the essential criteria for drug-likeness. Altogether, the findings reported in the current work clearly suggest that the identified anthranilamide derivatives have the potential to serve as effective drug candidates for future investigations.
Collapse
Affiliation(s)
- Sumera Zaib
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore 54590, Pakistan.
| | - Imtiaz Khan
- Department of Chemistry and Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester MI 7DN, UK.
| | - Hafiz Saqib Ali
- Chemistry Research Laboratory, Department of Chemistry, the INEOS Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, UK
| | - Muhammad Tayyab Younas
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore 54590, Pakistan
| | - Aliya Ibrar
- Department of Chemistry, Faculty of Physical and Applied Sciences, The University of Haripur, Haripur, KPK 22620, Pakistan.
| | - Abdel-Basit Al-Odayni
- Department of Restorative Dental Sciences, College of Dentistry, King Saud University, P.O. Box 60169, Riyadh 11545, Saudi Arabia
| | - Abdullah A Al-Kahtani
- Chemistry Department, College of Science, King Saud University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
35
|
Sanghai N, Vuong B, Burak Berk A, Afridi MSK, Tranmer GK. Current Small Molecule-Based Medicinal Chemistry Approaches for Neurodegeneration Therapeutics. ChemMedChem 2024; 19:e202300705. [PMID: 38329887 DOI: 10.1002/cmdc.202300705] [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: 12/14/2023] [Revised: 02/07/2024] [Accepted: 02/07/2024] [Indexed: 02/10/2024]
Abstract
Neurodegenerative diseases (NDDs) like Alzheimer's disease (AD), Parkinson's disease (PD), and Amyotrophic lateral sclerosis (ALS) possess multifactorial aetiologies. In recent years, our understanding of the biochemical and molecular pathways across NDDs has increased, however, new advances in small molecule-based therapeutic strategies targeting NDDs are obscure and scarce. Moreover, NDDs have been studied for more than five decades, however, there is a paucity of drugs that can treat NDDs. Further, the highly lipoidal blood-brain barrier (BBB) limits the uptake of many therapeutic molecules into the brain and is a complicating factor in the development of new agents to treat neurodegeneration. Considering the highly complex nature of NDDs, the association of multiple risk factors, and the challenges to overcome the BBB junction, medicinal chemists have developed small organic molecule-based novel approaches to target NDDs over the last few decades, such as designing lipophilic molecules and applying prodrug strategies. Attempts have been made to utilize a multitarget approach to modulate different biochemical molecular pathways involved in NDDs, in addition to, medicinal chemists making better decisions in identifying optimized drug candidates for the central nervous system (CNS) by using web-based computational tools. To increase the clinical success of these drug candidates, an in vitro assay modeling the BBB has been utilized by medicinal chemists in the pre-clinical phase as a further screening measure of small organic molecules. Herein, we examine some of the intriguing strategies taken by medicinal chemists to design small organic molecules to combat NDDs, with the intention of increasing our awareness of neurodegenerative therapeutics.
Collapse
Affiliation(s)
- Nitesh Sanghai
- College of Pharmacy, Rady Faculty of Health Science, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Billy Vuong
- College of Pharmacy, Rady Faculty of Health Science, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Ahmet Burak Berk
- College of Pharmacy, Rady Faculty of Health Science, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | | | - Geoffrey K Tranmer
- College of Pharmacy, Rady Faculty of Health Science, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| |
Collapse
|
36
|
Ujcikova H, Lee YS, Roubalova L, Svoboda P. The impact of multifunctional enkephalin analogs and morphine on the protein changes in crude membrane fractions isolated from the rat brain cortex and hippocampus. Peptides 2024; 174:171165. [PMID: 38307418 DOI: 10.1016/j.peptides.2024.171165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/12/2024] [Accepted: 01/29/2024] [Indexed: 02/04/2024]
Abstract
Endogenous opioid peptides serve as potent analgesics through the opioid receptor (OR) activation. However, they often suffer from poor metabolic stability, low lipophilicity, and low blood-brain barrier permeability. Researchers have developed many strategies to overcome the drawbacks of current pain medications and unwanted biological effects produced by the interaction with opioid receptors. Here, we tested multifunctional enkephalin analogs LYS739 (MOR/DOR agonist and KOR partial antagonist) and LYS744 (MOR/DOR agonist and KOR full antagonist) under in vivo conditions in comparison with MOR agonist, morphine. We applied 2D electrophoretic resolution to investigate differences in proteome profiles of crude membrane (CM) fractions isolated from the rat brain cortex and hippocampus exposed to the drugs (10 mg/kg, seven days). Our results have shown that treatment with analog LYS739 induced the most protein changes in cortical and hippocampal samples. The identified proteins were mainly associated with energy metabolism, cell shape and movement, apoptosis, protein folding, regulation of redox homeostasis, and signal transduction. Among these, the isoform of mitochondrial ATP synthase subunit beta (ATP5F1B) was the only protein upregulation in the hippocampus but not in the brain cortex. Contrarily, the administration of analog LYS744 caused a small number of protein alterations in both brain parts. Our results indicate that the KOR full antagonism, together with MOR/DOR agonism of multifunctional opioid ligands, can be beneficial in treating chronic pain states by reducing changes in protein expression levels but retaining analgesic efficacy.
Collapse
Affiliation(s)
- Hana Ujcikova
- Laboratory of Neurochemistry, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic.
| | - Yeon Sun Lee
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724, USA
| | - Lenka Roubalova
- Laboratory of Neurochemistry, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| | - Petr Svoboda
- Laboratory of Neurochemistry, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| |
Collapse
|
37
|
Reich N, Hölscher C. Cholecystokinin (CCK): a neuromodulator with therapeutic potential in Alzheimer's and Parkinson's disease. Front Neuroendocrinol 2024; 73:101122. [PMID: 38346453 DOI: 10.1016/j.yfrne.2024.101122] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/04/2024] [Accepted: 01/25/2024] [Indexed: 02/16/2024]
Abstract
Cholecystokinin (CCK) is a neuropeptide modulating digestion, glucose levels, neurotransmitters and memory. Recent studies suggest that CCK exhibits neuroprotective effects in Alzheimer's disease (AD) and Parkinson's disease (PD). Thus, we review the physiological function and therapeutic potential of CCK. The neuropeptide facilitates hippocampal glutamate release and gates GABAergic basket cell activity, which improves declarative memory acquisition, but inhibits consolidation. Cortical CCK alters recognition memory and enhances audio-visual processing. By stimulating CCK-1 receptors (CCK-1Rs), sulphated CCK-8 elicits dopamine release in the substantia nigra and striatum. In the mesolimbic pathway, CCK release is triggered by dopamine and terminates reward responses via CCK-2Rs. Importantly, activation of hippocampal and nigral CCK-2Rs is neuroprotective by evoking AMPK activation, expression of mitochondrial fusion modulators and autophagy. Other benefits include vagus nerve/CCK-1R-mediated expression of brain-derived neurotrophic factor, intestinal protection and suppression of inflammation. We also discuss caveats and the therapeutic combination of CCK with other peptide hormones.
Collapse
Affiliation(s)
- Niklas Reich
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, UK; Faculty of Health and Medicine, Biomedical & Life Sciences Division, Lancaster University, Lancaster LA1 4YQ, UK.
| | - Christian Hölscher
- Second associated Hospital, Neurology Department, Shanxi Medical University, Taiyuan, Shanxi, China; Henan Academy of Innovations in Medical Science, Neurodegeneration research group, Xinzhen, Henan province, China
| |
Collapse
|
38
|
Murumulla L, Bandaru LJM, Challa S. Heavy Metal Mediated Progressive Degeneration and Its Noxious Effects on Brain Microenvironment. Biol Trace Elem Res 2024; 202:1411-1427. [PMID: 37462849 DOI: 10.1007/s12011-023-03778-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/13/2023] [Indexed: 02/13/2024]
Abstract
Heavy metals, including lead (Pb), cadmium (Cd), arsenic (As), cobalt (Co), copper (Cu), manganese (Mn), zinc (Zn), and others, have a significant impact on the development and progression of neurodegenerative diseases in the human brain. This comprehensive review aims to consolidate the recent research on the harmful effects of different metals on specific brain cells such as neurons, microglia, astrocytes, and oligodendrocytes. Understanding the potential influence of these metals in neurodegeneration is crucial for effectively combating the ongoing advancement of these diseases. Metal-induced neurodegeneration involves molecular mechanisms such as apoptosis induction, dysregulation of metabolic and signaling pathways, metal imbalance, oxidative stress, loss of synaptic transmission, pathogenic peptide aggregation, and neuroinflammation. This review provides valuable insights by compiling the supportive evidence from recent research findings. Additionally, we briefly discuss the modes of action of natural neuroprotective compounds. While this comprehensive review aims to consolidate the recent research on the harmful effects of various metals on specific brain cells, it may not cover all studies and findings related to metal-induced neurodegeneration. Studies that are done using bioinformatics tools, microRNAs, long non-coding RNAs, emerging disease models, and studies based on the modes of exposure to toxic metals are a future prospect to be explored.
Collapse
Affiliation(s)
- Lokesh Murumulla
- Cell Biology Division, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad-500007, Hyderabad, Telangana, India
| | - Lakshmi Jaya Madhuri Bandaru
- Cell Biology Division, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad-500007, Hyderabad, Telangana, India
| | - Suresh Challa
- Cell Biology Division, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad-500007, Hyderabad, Telangana, India.
| |
Collapse
|
39
|
Cao H, Li B, Mu M, Li S, Chen H, Tao H, Wang W, Zou Y, Zhao Y, Liu Y, Tao X. Nicotine suppresses crystalline silica-induced astrocyte activation and neuronal death by inhibiting NF-κB in the mouse hippocampus. CNS Neurosci Ther 2024; 30:e14508. [PMID: 37864452 PMCID: PMC11017465 DOI: 10.1111/cns.14508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/25/2023] [Accepted: 10/06/2023] [Indexed: 10/22/2023] Open
Abstract
AIMS Exposure to crystalline silica (CS) in occupational settings induces chronic inflammation in the respiratory system and, potentially, the brain. Some workers are frequently concurrently exposed to both CS and nicotine. Here, we explored the impact of nicotine on CS-induced neuroinflammation in the mouse hippocampus. METHODS In this study, we established double-exposed models of CS and nicotine in C57BL/6 mice. To assess depression-like behavior, experiments were conducted at 3, 6, and 9 weeks. Serum inflammatory factors were analyzed by ELISA. Hippocampus was collected for RNA sequencing analysis and examining the gene expression patterns linked to inflammation and cell death. Microglia and astrocyte activation and hippocampal neuronal death were assessed using immunohistochemistry and immunofluorescence staining. Western blotting was used to analyze the NF-κB expression level. RESULTS Mice exposed to CS for 3 weeks showed signs of depression. This was accompanied by elevated IL-6 in blood, destruction of the blood-brain barrier, and activation of astrocytes caused by an increased NF-κB expression in the CA1 area of the hippocampus. The elevated levels of astrocyte-derived Lcn2 and upregulated genes related to inflammation led to higher neuronal mortality. Moreover, nicotine mitigated the NF-κB expression, astrocyte activation, and neuronal death, thereby ameliorating the associated symptoms. CONCLUSION Silica exposure induces neuroinflammation and neuronal death in the mouse hippocampal CA1 region and depressive behavior. However, nicotine inhibits CS-induced neuroinflammation and neuronal apoptosis, alleviating depressive-like behaviors in mice.
Collapse
Affiliation(s)
- Hangbing Cao
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Bing Li
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Min Mu
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Shanshan Li
- School of PharmacyBengbu Medical CollegeBengbuChina
| | - Haoming Chen
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Huihui Tao
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Wenyang Wang
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Yuanjie Zou
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Yehong Zhao
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Yang Liu
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Xinrong Tao
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| |
Collapse
|
40
|
Amro Z, Collins-Praino L, Yool A. Protective roles of peroxiporins AQP0 and AQP11 in human astrocyte and neuronal cell lines in response to oxidative and inflammatory stressors. Biosci Rep 2024; 44:BSR20231725. [PMID: 38451099 PMCID: PMC10965398 DOI: 10.1042/bsr20231725] [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] [Revised: 03/03/2024] [Accepted: 03/06/2024] [Indexed: 03/08/2024] Open
Abstract
In addition to aquaporin (AQP) classes AQP1, AQP4 and AQP9 known to be expressed in mammalian brain, our recent transcriptomic analyses identified AQP0 and AQP11 in human cortex and hippocampus at levels correlated with age and Alzheimer's disease (AD) status; however, protein localization remained unknown. Roles of AQP0 and AQP11 in transporting hydrogen peroxide (H2O2) in lens and kidney prompted our hypothesis that up-regulation in brain might similarly be protective. Established cell lines for astroglia (1321N1) and neurons (SHSY5Y, differentiated with retinoic acid) were used to monitor changes in transcript levels for human AQPs (AQP0 to AQP12) in response to inflammation (simulated with 10-100 ng/ml lipopolysaccharide [LPS], 24 h), and hypoxia (5 min N2, followed by 0 to 24 h normoxia). AQP transcripts up-regulated in both 1321N1 and SHSY5Y included AQP0, AQP1 and AQP11. Immunocytochemistry in 1321N1 cells confirmed protein expression for AQP0 and AQP11 in plasma membrane and endoplasmic reticulum; AQP11 increased 10-fold after LPS and AQP0 increased 0.3-fold. In SHSY5Y cells, AQP0 expression increased 0.2-fold after 24 h LPS; AQP11 showed no appreciable change. Proposed peroxiporin roles were tested using melondialdehyde (MDA) assays to quantify lipid peroxidation levels after brief H2O2. Boosting peroxiporin expression by LPS pretreatment lowered subsequent H2O2-induced MDA responses (∼50%) compared with controls; conversely small interfering RNA knockdown of AQP0 in 1321N1 increased lipid peroxidation (∼17%) after H2O2, with a similar trend for AQP11 siRNA. Interventions that increase native brain peroxiporin activity are promising as new approaches to mitigate damage caused by aging and neurodegeneration.
Collapse
Affiliation(s)
- Zein Amro
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia
| | | | - Andrea J. Yool
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
41
|
Beltrán-Velasco AI, Reiriz M, Uceda S, Echeverry-Alzate V. Lactiplantibacillus (Lactobacillus) plantarum as a Complementary Treatment to Improve Symptomatology in Neurodegenerative Disease: A Systematic Review of Open Access Literature. Int J Mol Sci 2024; 25:3010. [PMID: 38474254 PMCID: PMC10931784 DOI: 10.3390/ijms25053010] [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: 12/14/2023] [Revised: 02/28/2024] [Accepted: 03/03/2024] [Indexed: 03/14/2024] Open
Abstract
This systematic review addresses the use of Lactiplantibacillus (Lactobacillus) plantarum in the symptomatological intervention of neurodegenerative disease. The existence of gut microbiota dysbiosis has been associated with systemic inflammatory processes present in neurodegenerative disease, creating the opportunity for new treatment strategies. This involves modifying the strains that constitute the gut microbiota to enhance synaptic function through the gut-brain axis. Recent studies have evaluated the beneficial effects of the use of Lactiplantibacillus plantarum on motor and cognitive symptomatology, alone or in combination. This systematic review includes 20 research articles (n = 3 in human and n = 17 in animal models). The main result of this research was that the use of Lactiplantibacillus plantarum alone or in combination produced improvements in symptomatology related to neurodegenerative disease. However, one of the studies included reported negative effects after the administration of Lactiplantibacillus plantarum. This systematic review provides current and relevant information about the use of this probiotic in pathologies that present neurodegenerative processes such as Alzheimer's disease, Parkinson's disease and Multiple Sclerosis.
Collapse
Affiliation(s)
| | | | - Sara Uceda
- Psychology Department, School of Life and Nature Sciences, Nebrija University, 28240 Madrid, Spain; (A.I.B.-V.); (M.R.)
| | - Víctor Echeverry-Alzate
- Psychology Department, School of Life and Nature Sciences, Nebrija University, 28240 Madrid, Spain; (A.I.B.-V.); (M.R.)
| |
Collapse
|
42
|
Harrison MAA, Morris SL, Rudman GA, Rittenhouse DJ, Monk CH, Sakamuri SSVP, Mehedi Hasan M, Shamima Khatun M, Wang H, Garfinkel LP, Norton EB, Kim S, Kolls JK, Jazwinski SM, Mostany R, Katakam PVG, Engler-Chiurazzi EB, Zwezdaryk KJ. Intermittent cytomegalovirus infection alters neurobiological metabolism and induces cognitive deficits in mice. Brain Behav Immun 2024; 117:36-50. [PMID: 38182037 PMCID: PMC11914963 DOI: 10.1016/j.bbi.2023.12.033] [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: 07/24/2023] [Revised: 12/21/2023] [Accepted: 12/23/2023] [Indexed: 01/07/2024] Open
Abstract
Risk factors contributing to dementia are multifactorial. Accumulating evidence suggests a role for pathogens as risk factors, but data is largely correlative with few causal relationships. Here, we demonstrate that intermittent murine cytomegalovirus (MCMV) infection of mice, alters blood brain barrier (BBB) permeability and metabolic pathways. Increased basal mitochondrial function is observed in brain microvessels cells (BMV) exposed to intermittent MCMV infection and is accompanied by elevated levels of superoxide. Further, mice score lower in cognitive assays compared to age-matched controls who were never administered MCMV. Our data show that repeated systemic infection with MCMV, increases markers of neuroinflammation, alters mitochondrial function, increases markers of oxidative stress and impacts cognition. Together, this suggests that viral burden may be a risk factor for dementia. These observations provide possible mechanistic insights through which pathogens may contribute to the progression or exacerbation of dementia.
Collapse
Affiliation(s)
- Mark A A Harrison
- Neuroscience Program, Tulane Brain Institute, Tulane University School of Science & Engineering, New Orleans, LA 70112, USA; Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Sara L Morris
- Biomedical Sciences Program, Tulane University School of Medicine, New Orleans, LA 70112, USA; Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Grace A Rudman
- Department of Environmental Studies, Tulane University School of Liberal Arts, New Orleans, LA 70112, USA
| | - Daniel J Rittenhouse
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA; Tulane Center for Translational Research in Infection & Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Chandler H Monk
- Bioinnovation Program, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Siva S V P Sakamuri
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Md Mehedi Hasan
- Tulane Center for Aging, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Mst Shamima Khatun
- Tulane Center for Translational Research in Infection & Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Hanyun Wang
- Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Lucas P Garfinkel
- Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Elizabeth B Norton
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Sangku Kim
- Tulane Center for Aging, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jay K Kolls
- Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA; Tulane Center for Translational Research in Infection & Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - S. Michal Jazwinski
- Tulane Center for Aging, Tulane University School of Medicine, New Orleans, LA 70112, USA; Tulane Brain Institute, Tulane University School of Medicine, New Orleans, LA 70112, USA; Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Ricardo Mostany
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA; Tulane Center for Aging, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Prasad V G Katakam
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA; Tulane Center for Aging, Tulane University School of Medicine, New Orleans, LA 70112, USA; Tulane Brain Institute, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Elizabeth B Engler-Chiurazzi
- Tulane Center for Aging, Tulane University School of Medicine, New Orleans, LA 70112, USA; Tulane Brain Institute, Tulane University School of Medicine, New Orleans, LA 70112, USA; Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| | - Kevin J Zwezdaryk
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA; Tulane Center for Aging, Tulane University School of Medicine, New Orleans, LA 70112, USA; Tulane Brain Institute, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| |
Collapse
|
43
|
Davidson TL, Stevenson RJ. Vulnerability of the Hippocampus to Insults: Links to Blood-Brain Barrier Dysfunction. Int J Mol Sci 2024; 25:1991. [PMID: 38396670 PMCID: PMC10888241 DOI: 10.3390/ijms25041991] [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: 01/03/2024] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
The hippocampus is a critical brain substrate for learning and memory; events that harm the hippocampus can seriously impair mental and behavioral functioning. Hippocampal pathophysiologies have been identified as potential causes and effects of a remarkably diverse array of medical diseases, psychological disorders, and environmental sources of damage. It may be that the hippocampus is more vulnerable than other brain areas to insults that are related to these conditions. One purpose of this review is to assess the vulnerability of the hippocampus to the most prevalent types of insults in multiple biomedical domains (i.e., neuroactive pathogens, neurotoxins, neurological conditions, trauma, aging, neurodegenerative disease, acquired brain injury, mental health conditions, endocrine disorders, developmental disabilities, nutrition) and to evaluate whether these insults affect the hippocampus first and more prominently compared to other brain loci. A second purpose is to consider the role of hippocampal blood-brain barrier (BBB) breakdown in either causing or worsening the harmful effects of each insult. Recent research suggests that the hippocampal BBB is more fragile compared to other brain areas and may also be more prone to the disruption of the transport mechanisms that act to maintain the internal milieu. Moreover, a compromised BBB could be a factor that is common to many different types of insults. Our analysis indicates that the hippocampus is more vulnerable to insults compared to other parts of the brain, and that developing interventions that protect the hippocampal BBB may help to prevent or ameliorate the harmful effects of many insults on memory and cognition.
Collapse
Affiliation(s)
- Terry L. Davidson
- Department of Neuroscience, Center for Neuroscience and Behavior, American University, 4400 Massachusetts Avenue, NW, Washington, DC 20016, USA
| | | |
Collapse
|
44
|
Rossi C, Distaso M, Raggi F, Kusmic C, Faita F, Solini A. Lacking P2X7-receptors protects substantia nigra dopaminergic neurons and hippocampal-related cognitive performance from the deleterious effects of high-fat diet exposure in adult male mice. Front Nutr 2024; 11:1289750. [PMID: 38344021 PMCID: PMC10854005 DOI: 10.3389/fnut.2024.1289750] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 01/11/2024] [Indexed: 01/05/2025] Open
Abstract
BACKGROUND Dietary fat consumption, involved in the pathogenesis of insulin resistance and impaired glucose metabolism, is linked with decline in cognitive functions, dementia, and development of Parkinson's disease and Alzheimer's disease. Mature IL-1β, requiring the activation of the P2X7 receptor (P2X7R)-inflammasome complex, is an important mediator of neuroinflammation. The aim of the study was to test whether P2X7R activation might interfere with systemic and cerebral metabolic homeostasis. METHODS We treated WT and P2X7R KO mice with a high-fat diet (HFD) for 16 weeks, evaluating the effects on the Substantia Nigra and Hippocampus, target areas of damage in several forms of cognitive impairment. RESULTS HFD-treated WT and P2X7R KO mice showed a different brain mRNA profile of Insulin and Igf-1, with these genes and relative receptors, more expressed in KO mice. Unlike P2X7R KO mice, WT mice treated with HFD displayed a diameter reduction in dopaminergic neurons in the Substantia Nigra, accompanied by an increased IBA1 expression in this area; they also showed poor performances during Y-Maze and Morris Water Maze, tasks involving Hippocampus activity. Conversely, Parkin, whose reduction might promote neuronal cell death, was increased in the brain of P2X7R KO animals. CONCLUSION We report for the first time that HFD induces damage in dopaminergic neurons of the Substantia Nigra and a Hippocampus-related worse cognitive performance, both attenuated in the absence of P2X7R. The involved mechanisms might differ in the two brain areas, with a predominant role of inflammation in the Substantia Nigra and a metabolic derangement in the Hippocampus.
Collapse
Affiliation(s)
- Chiara Rossi
- Department of Surgical, Medical, Molecular and Critical Area Pathology University of Pisa, Pisa, Italy
| | - Mariarosaria Distaso
- Department of Surgical, Medical, Molecular and Critical Area Pathology University of Pisa, Pisa, Italy
| | - Francesco Raggi
- Department of Surgical, Medical, Molecular and Critical Area Pathology University of Pisa, Pisa, Italy
| | | | | | - Anna Solini
- Department of Surgical, Medical, Molecular and Critical Area Pathology University of Pisa, Pisa, Italy
| |
Collapse
|
45
|
Skibska A, Perlikowska R. Natural Plant Materials as a Source of Neuroprotective Peptides. Curr Med Chem 2024; 31:5027-5045. [PMID: 37403392 DOI: 10.2174/0929867331666230703145043] [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: 03/30/2023] [Revised: 05/24/2023] [Accepted: 06/01/2023] [Indexed: 07/06/2023]
Abstract
In many circumstances, some crucial elements of the neuronal defense system fail, slowly leading to neurodegenerative diseases. Activating this natural process by administering exogenous agents to counteract unfavourable changes seems promising. Therefore, looking for neuroprotective therapeutics, we have to focus on compounds that inhibit the primary mechanisms leading to neuronal injuries, e.g., apoptosis, excitotoxicity, oxidative stress, and inflammation. Among many compounds considered neuroprotective agents, protein hydrolysates and peptides derived from natural materials or their synthetic analogues are good candidates. They have several advantages, such as high selectivity and biological activity, a broad range of targets, and high safety profile. This review aims to provide biological activities, the mechanism of action and the functional properties of plant-derived protein hydrolysates and peptides. We focused on their significant role in human health by affecting the nervous system and having neuroprotective and brain-boosting properties, leading to memory and cognitive improving activities. We hope our observation may guide the evaluation of novel peptides with potential neuroprotective effects. Research into neuroprotective peptides may find application in different sectors as ingredients in functional foods or pharmaceuticals to improve human health and prevent diseases.
Collapse
Affiliation(s)
- Agnieszka Skibska
- Department of Biomolecular Chemistry, Faculty of Medicine, Medical University, Lodz, Poland
| | - Renata Perlikowska
- Department of Biomolecular Chemistry, Faculty of Medicine, Medical University, Lodz, Poland
| |
Collapse
|
46
|
Ye YC, Chai SF, Li XR, Wu MN, Cai HY, Wang ZJ. Intermittent fasting and Alzheimer's disease-Targeting ketone bodies as a potential strategy for brain energy rescue. Metab Brain Dis 2024; 39:129-146. [PMID: 37823968 DOI: 10.1007/s11011-023-01288-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 09/01/2023] [Indexed: 10/13/2023]
Abstract
Alzheimer's disease (AD) lacks effective clinical treatments. As the disease progresses, the cerebral glucose hypometabolism that appears in the preclinical phase of AD gradually worsens, leading to increasingly severe brain energy disorders. This review analyzes the brain energy deficit in AD and its etiology, brain energy rescue strategies based on ketone intervention, the effects and mechanisms of IF, the differences in efficacy between IF and ketogenic diet and the duality of IF. The evidence suggests that brain energy deficits lead to the development and progression of AD pathology. IF, which improves brain energy impairments by promoting ketone metabolism, thus has good therapeutic potential for AD.
Collapse
Affiliation(s)
- Yu- Cai Ye
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Shi-Fan Chai
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Xin-Ru Li
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Mei-Na Wu
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Hong-Yan Cai
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Zhao-Jun Wang
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China.
| |
Collapse
|
47
|
Samanta S, Chakraborty S, Bagchi D. Pathogenesis of Neurodegenerative Diseases and the Protective Role of Natural Bioactive Components. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2024; 43:20-32. [PMID: 37186678 DOI: 10.1080/27697061.2023.2203235] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023]
Abstract
Neurodegenerative diseases are a serious problem throughout the world. There are several causes of neurodegenerative diseases; these include genetic predisposition, accumulation of misfolded proteins, oxidative stress, neuroinflammation, and excitotoxicity. Oxidative stress increases the production of reactive oxygen species (ROS) that advance lipid peroxidation, DNA damage, and neuroinflammation. The cellular antioxidant system (superoxide dismutase, catalase, peroxidase, and reduced glutathione) plays a crucial role in scavenging free radicals. An imbalance in the defensive actions of antioxidants and overproduction of ROS intensify neurodegeneration. The formation of misfolded proteins, glutamate toxicity, oxidative stress, and cytokine imbalance promote the pathogenesis of Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. Antioxidants are now attractive molecules to fight against neurodegeneration. Certain vitamins (A, E, C) and polyphenolic compounds (flavonoids) show excellent antioxidant properties. Diet is the major source of antioxidants. However, diet medicinal herbs are also rich sources of numerous flavonoids. Antioxidants prevent ROS-mediated neuronal degeneration in post-oxidative stress conditions. The present review is focused on the pathogenesis of neurodegenerative diseases and the protective role of antioxidants. KEY TEACHING POINTSThis review shows that multiple factors are directly or indirectly associated with the pathogenesis of neurodegenerative diseases.Failure to cellular antioxidant capacity increases oxidative stress that intensifies neuroinflammation and disease progression.Different vitamins, carotenoids, and flavonoids, having antioxidant capacity, can be considered protective agents.
Collapse
Affiliation(s)
- Saptadip Samanta
- Department of Physiology, Midnapore College, Midnapore, West Bengal, India
| | - Sanjoy Chakraborty
- Department of Biological Sciences, New York City College of Technology/CUNY, Brooklyn, New York, USA
| | - Debasis Bagchi
- Department of Biology, College of Arts and Sciences, Adelphi University, Garden City, New York, USA
- Department of Psychology, Gordon F. Derner School of Psychology, Adelphi University, Garden City, New York, USA
| |
Collapse
|
48
|
Czuczwar SJ, Kocki J, Miziak B, Bogucki J, Bogucka-Kocka A, Pluta R. Alpha-, Beta-, and Gamma-Secretase, Amyloid Precursor Protein, and Tau Protein Genes in the Hippocampal CA3 Subfield in an Ischemic Model of Alzheimer's Disease with Survival up to 2 Years. J Alzheimers Dis 2024; 98:151-161. [PMID: 38393914 PMCID: PMC10977426 DOI: 10.3233/jad-231333] [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] [Accepted: 12/26/2023] [Indexed: 02/25/2024]
Abstract
Background Understanding the phenomena underlying the non-selective susceptibility to ischemia of pyramidal neurons in the CA3 is important from the point of view of elucidating the mechanisms of memory loss and the development of dementia. Objective The aim of the study was to investigate changes in genes expression of amyloid precursor protein, its cleaving enzymes and tau protein in CA3 post-ischemia with survival of 12-24 months. Methods We used an ischemic model of Alzheimer's disease to study the above genes using an RT-PCR protocol. Results The expression of the amyloid precursor protein gene was above the control values at all times post-ischemia. The expression of the α-secretase gene also exceeded the control values post-ischemia. The expression of the β-secretase gene increased 12 and 24 months post-ischemia, and 18 months was below control values. Presenilin 1 and 2 genes expression was significantly elevated at all times post-ischemia. Also, tau protein gene expression was significantly elevated throughout the observation period, and peak gene expression was present 12 months post-ischemia. Conclusions The study suggests that the genes studied are involved in the non-amyloidogenic processing of amyloid precursor protein. Additionally data indicate that brain ischemia with long-term survival causes damage and death of pyramidal neurons in the CA3 area of the hippocampus in a modified tau protein-dependent manner. Thus defining a new and important mechanism of pyramidal neuronal death in the CA3 area post-ischemia. In addition expression of tau protein gene modification after brain ischemia is useful in identifying ischemic mechanisms occurring in Alzheimer's disease.
Collapse
Affiliation(s)
| | - Janusz Kocki
- Department of Clinical Genetics, Medical University of Lublin, Lublin, Poland
| | - Barbara Miziak
- Department of Pathophysiology, Medical University of Lublin, Lublin, Poland
| | - Jacek Bogucki
- Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Anna Bogucka-Kocka
- Department of Biology and Genetics, Medical University of Lublin, Lublin, Poland
| | - Ryszard Pluta
- Department of Pathophysiology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
49
|
Dutta D, Tripathi D, Asthana P, Rana K, Jain N, Sharma R, Naithani U, Jauhari D, Rachana. Therapeutic Effects of Capsaicin on Central Nervous Disorders with Special Emphasis on Parkinson’s and Alzheimer’s Diseases. CAPSAICINOIDS 2024:489-510. [DOI: 10.1007/978-981-99-7779-6_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
50
|
Sabry K, Jamshidi Z, Emami SA, Sahebka A. Potential therapeutic effects of baicalin and baicalein. AVICENNA JOURNAL OF PHYTOMEDICINE 2024; 14:23-49. [PMID: 38948180 PMCID: PMC11210699 DOI: 10.22038/ajp.2023.22307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/24/2023] [Accepted: 01/24/2024] [Indexed: 07/02/2024]
Abstract
Objective Baicalin and baicalein are natural flavonoids reported for the first time from Scutellaria baicalensis Georgi. Recently, attention has been paid to these valuable flavonoids due to their promising effects. This paper aims to have a comprehensive review of their pharmacological effects. Materials and Methods An extensive search through scientific databases including Scopus, PubMed, and ISI Web of Science was established. Results According to literature, these compounds have been mainly effective in the treatment of neurological and neurodegenerative diseases, hepatic and cardiovascular disorders, metabolic syndrome, and cancers through anti-inflammatory and antioxidant pathways. Induction of apoptosis and autophagy, and inhibition of migration and metastasis are the main mechanisms for their cytotoxic and antitumor activities. Decreasing inflammation, reducing oxidative stress, regulating the metabolism of lipids, and decreasing fibrosis, apoptosis, and steatosis are their main hepatoprotective mechanisms. Inhibiting the development of cardiac fibrosis and reducing inflammation, oxidative stress, and apoptosis are also the mechanisms suggested for cardioprotective activities. Decreasing the accumulation of inflammatory mediators and improving cognitive function and depressive-like behaviours are the main mechanisms for neurological and neurodegenerative activities. Conclusion The findings suggest the therapeutic potential of baicalin and baicalein. However, complementary research in different in vitro and in vivo models to investigate their mechanisms of action as well as clinical trials to evaluate their efficacy and safety are suggested.
Collapse
Affiliation(s)
- Kamyar Sabry
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Jamshidi
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Ahmad Emami
- Department of Traditional Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebka
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|