51
|
Uno H, Itokazu T, Yamashita T. Inhibition of repulsive guidance molecule A ameliorates diabetes-induced cognitive decline and hippocampal neurogenesis impairment in mice. Commun Biol 2025; 8:263. [PMID: 39972167 PMCID: PMC11840113 DOI: 10.1038/s42003-025-07696-7] [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: 05/30/2024] [Accepted: 02/06/2025] [Indexed: 02/21/2025] Open
Abstract
Although diabetes mellitus is strongly associated with dementia, the mechanism underlying diabetes-induced cognitive dysfunction has not been clarified. Here, we demonstrate the vital role of repulsive guidance molecule A (RGMa) in the regulation of adult hippocampal neurogenesis and cognitive impairment under diabetic conditions. In type 2 diabetic db/db mice and streptozotocin-mediated type 1 diabetic mice, RGMa is upregulated in the granular cell layer of the dentate gyrus. Additionally, both neural stem cells (NSCs) and immature neurons express its receptor, neogenin. In vitro experiments revealed that high glucose-conditioned hippocampal neurons inhibited the differentiation of NSCs, and the application of an anti-RGMa antibody restored it. The treatment with an anti-RGMa antibody ameliorated diabetes-induced cognitive decline and impairment of hippocampal neurogenesis. These findings suggest that the RGMa negatively regulates hippocampal neurogenesis and is involved in diabetes mellitus-induced cognitive decline.
Collapse
Affiliation(s)
- Hiroki Uno
- Department of Molecular Neurosciences, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Takahide Itokazu
- Department of Molecular Neurosciences, Graduate School of Medicine, Osaka University, Suita, Japan.
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Japan.
| | - Toshihide Yamashita
- Department of Molecular Neurosciences, Graduate School of Medicine, Osaka University, Suita, Japan.
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Japan.
- WPI-Immunology Frontier Research Center, Osaka University, Suita, Japan.
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan.
| |
Collapse
|
52
|
Xiang Q, Lin H, Tao JS, Fu CJ, Liu LN, Deng J, Li XH. MiR-18a-LncRNA NONRATG-022419 pairs targeted PRG-1 regulates diabetic induced cognitive impairment by regulating NGF\BDNF-Trkb signaling pathway. Proteome Sci 2025; 23:1. [PMID: 39915794 PMCID: PMC11800523 DOI: 10.1186/s12953-025-00239-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 01/06/2025] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND Diabetic encephalopathy (DE) is considered as one of the complications of diabetes,which is associated with cognitive impairment in the pathological process of development. Up to now, phospholipid phosphatase related 4 (Plppr4), also known as plasticity related gene 1 (PRG-1) has been revealed its important role in neuroplasticity. However, the underlying mechanisms of Plppr4 on the basis of diabetic-induced cognitive dysfunction (DCD) are still unknown. The aim of current study was to provide insight into molecular mechanism and cellular heterogeneity underlying DCD, and investigate the functional role of PRG-1 involved in this process. METHODS Combined Single-cell RNA sequencing (scRNA-seq) and RNA transcriptome analysis, the distinct sub-populations, functional heterogeneity as well as potential enriched signaling pathways of hippocampal cells could be elucidated. RESULTS We identified the sub-cluster of type I spiral ganglion neurons expressed marker gene as Amigo2 in cluster8 and Cnr1 in cluster 9 of hippocampal cells from DCD and the effect of those on neuronal cells interaction. We also found that PRG-1 was involved in the synaptic plasticity regulation of hippocampus via NGF\BDNF-Trkb signaling pathway. In high glucose induced HT22 cells injury model in vitro, we investigated that down-regulated PRG-1 along with down-regulated BDNF and also decreased expression of synapsin-1, PSD-95, SYN which are related to synaptic plasticity; Meanwhile, the Prg-1 targeted miR-18a-LncRNA NONRATG-022419 pairs related with significantly down-regulated expression of PRG-1. CONCLUSION This study revealed the synaptic plasticity regulation of PRG-1 in DCD, and might provide the therapeutic target and potential biomarkers for early interventions in DCD patients.
Collapse
Affiliation(s)
- Qiong Xiang
- Institute of Medicine, Medical Research Center, Jishou University, Hunan, China
| | - Hu Lin
- Institute of Medicine, Jishou University, Ren min south road 120#, Jishou, Hunan, 416000, China
| | - Jia-Sheng Tao
- Institute of Medicine, Medical Research Center, Jishou University, Hunan, China
| | - Chuan-Jun Fu
- Institute of Medicine, Medical Research Center, Jishou University, Hunan, China
| | - Li-Ni Liu
- Institute of Medicine, Medical Research Center, Jishou University, Hunan, China
| | - Jing Deng
- Institute of Medicine, Medical Research Center, Jishou University, Hunan, China
| | - Xian-Hui Li
- Institute of Medicine, Jishou University, Ren min south road 120#, Jishou, Hunan, 416000, China.
| |
Collapse
|
53
|
Shilikbay T, Nawaz A, Doon M, Ceman S. RNA helicase MOV10 suppresses fear memory and dendritic arborization and regulates microtubule dynamics in hippocampal neurons. BMC Biol 2025; 23:36. [PMID: 39915816 PMCID: PMC11803958 DOI: 10.1186/s12915-025-02138-6] [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: 01/30/2024] [Accepted: 01/21/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND RNA helicase MOV10 is highly expressed in postnatal brain and associates with FMRP and AGO2, suggesting a role in translation regulation in learning and memory. RESULTS We generated a brain-specific knockout mouse (Mov10 Deletion) with greatly reduced MOV10 expression in cortex and hippocampus. Behavior testing revealed enhanced fear memory, similar to that observed in a mouse with reduced brain microRNA production, supporting MOV10's reported role as an AGO2 cofactor. Cultured hippocampal neurons have elongated distal dendrites, a reported feature of augmin/HAUS over-expression in Drosophila da sensory neurons. In mitotic spindle formation, HAUS is antagonized by the microtubule bundling protein NUMA1. Numa1 mRNA is a MOV10 CLIP target and is among the genes significantly decreased in Mov10 Deletion hippocampus. Restoration of NUMA1 expression and knockdown of HAUS rescued phenotypes of the Mov10 Deletion hippocampal neurons. CONCLUSIONS This is the first evidence of translation regulation of NUMA1 by MOV10 as a control point in dendritogenesis.
Collapse
Affiliation(s)
- Temirlan Shilikbay
- Cell and Developmental Biology, University of Illinois Urbana-Champaign, Urbana, USA
| | - Aatiqa Nawaz
- Cell and Developmental Biology, University of Illinois Urbana-Champaign, Urbana, USA
| | - Megan Doon
- Molecular and Cellular Biology, University of Illinois Urbana-Champaign, Urbana, USA
| | - Stephanie Ceman
- Cell and Developmental Biology, University of Illinois Urbana-Champaign, Urbana, USA.
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
54
|
Queen NJ, Zou X, Huang W, Mohammed T, Cao L. Environmental Enrichment Normalizes Metabolic Function in the Murine Model of Prader-Willi Syndrome Magel2-Null Mice. Endocrinology 2025; 166:bqaf001. [PMID: 39801003 PMCID: PMC11808065 DOI: 10.1210/endocr/bqaf001] [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: 11/04/2024] [Indexed: 02/11/2025]
Abstract
Prader-Willi syndrome (PWS) is a rare genetic disease that causes developmental delays, intellectual impairment, constant hunger, obesity, endocrine dysfunction, and various behavioral and neuropsychiatric abnormalities. Standard care of PWS is limited to strict supervision of food intake and GH therapy, highlighting the unmet need for new therapeutic strategies. Environmental enrichment (EE), a housing environment providing physical, social, and cognitive stimulations, exerts broad benefits on mental and physical health. Here we assessed the metabolic and behavioral effects of EE in the Magel2-null mouse model of PWS. EE initiated after the occurrence of metabolic abnormality was sufficient to normalize body weight and body composition, reverse hyperleptinemia, and improve glucose metabolism in the male Magel2-null mice. These metabolic improvements induced by EE were comparable to those achieved by a hypothalamic brain-derived neurotrophic factor gene therapy although the underlying mechanisms remain to be determined. These data suggest biobehavioral interventions such as EE could be effective in the treatment of PWS-related metabolic abnormalities.
Collapse
Affiliation(s)
- Nicholas J Queen
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Xunchang Zou
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Wei Huang
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Tawfiq Mohammed
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Lei Cao
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
55
|
Lahogue C, Boulouard M, Menager F, Freret T, Billard JM, Bouet V. A new 2-hit model combining serine racemase deletion and maternal separation displays behavioral and cognitive deficits associated with schizophrenia. Behav Brain Res 2025; 477:115301. [PMID: 39442565 DOI: 10.1016/j.bbr.2024.115301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Schizophrenia (SCZ) is a multifactorial psychotic disorder characterized by positive and negative symptoms as well as cognitive impairments. To advance the current treatments, it is important to improve animal models by considering the multifactorial etiology, thus by combining different risk factors. The objective of our study was to explore in a new mouse model, the impact of genetic deletion of serine racemase (genetic vulnerability) combined with an early stress factor induced by maternal separation (early environmental exposure) in the context of SCZ development. The face validity of the model was assessed through a wide range of behavioral experiments. The 2-hit mice displayed an increased locomotor activity mimicking positive symptoms, working memory impairment, cognitive deficits and recognition memory alterations, which could reflect neophobia. This new multifactorial model therefore presents an interesting phenotype for modelling animal model with partial behavioral and cognitive deficits associated with SCZ.
Collapse
Affiliation(s)
- Caroline Lahogue
- Normandie Univ, UNICAEN, INSERM, FHU A2M2P, COMETE, Caen 14000, France.
| | - Michel Boulouard
- Normandie Univ, UNICAEN, INSERM, FHU A2M2P, COMETE, Caen 14000, France
| | - François Menager
- Normandie Univ, UNICAEN, INSERM, FHU A2M2P, COMETE, Caen 14000, France
| | - Thomas Freret
- Normandie Univ, UNICAEN, INSERM, FHU A2M2P, COMETE, Caen 14000, France
| | | | - Valentine Bouet
- Normandie Univ, UNICAEN, INSERM, FHU A2M2P, COMETE, Caen 14000, France.
| |
Collapse
|
56
|
Guareschi F, Fonseca C, Silva S, Pescina S, Nicoli S, Buttini F, Sonvico F, Fortuna A. Therapeutic effect of cyclosporine A-loading TPGS micelles on a mouse model of LPS-induced neuroinflammation. Eur J Pharm Sci 2025; 205:106994. [PMID: 39701548 DOI: 10.1016/j.ejps.2024.106994] [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: 10/11/2024] [Revised: 12/07/2024] [Accepted: 12/17/2024] [Indexed: 12/21/2024]
Abstract
Neuroinflammation is an undoubted hallmark of neurodegenerative processes characterized by memory impairment, loss of coordination and muscle strength in diseases such as Alzheimer's disease, Parkinson's disease and multiple sclerosis as well as depressive disorders. Cyclosporine A (CSA) has already been identified as a promising neuroprotective peptide, due to its well-known anti-inflammatory properties. Herein, CSA was encapsulated into α-tocopherol polyethylene glycol 1000 succinate (TPGS) micelles and intranasally administered to a lipopolysaccharide (LPS) induced mouse model of neuroinflammation. After the treatment, mice were subjected to behavioral tests to assess cognitive and motor skills, while the biodistribution of CSA in plasma and olfactory bulb was studied by a new HPLC method validated for precision and accuracy. The results highlighted that in comparison to the classic oral CSA suspension, the intranasal (IN) administration showed significatively better safety and efficiency profiles. Notably, IN administration of CSA micelles showed relevant antidepressive effects and a certain ability to revert LPS-induced motor impairment. This work pointed out that the innovative and noninvasive IN administration of TPGS micelles could represent a safe and effective alternative to the classic oral route to deliver CSA at the Central Nervous System level, where its beneficial activity against neuroinflammation can be exploited.
Collapse
Affiliation(s)
- Fabiola Guareschi
- Advanced Drug Delivery Research Laboratory, Department of Food and Drug Science, University of Parma, Parma, Italy
| | - Carla Fonseca
- Laboratory of Pharmacology, Department of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT/ICNAS - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal; Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain; Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - Soraia Silva
- Laboratory of Pharmacology, Department of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT/ICNAS - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Silvia Pescina
- Advanced Drug Delivery Research Laboratory, Department of Food and Drug Science, University of Parma, Parma, Italy
| | - Sara Nicoli
- Advanced Drug Delivery Research Laboratory, Department of Food and Drug Science, University of Parma, Parma, Italy
| | - Francesca Buttini
- Advanced Drug Delivery Research Laboratory, Department of Food and Drug Science, University of Parma, Parma, Italy; University Research Centre for the Innovation of Health Products, Biopharmanet-TEC, University of Parma, Parma, Italy
| | - Fabio Sonvico
- Advanced Drug Delivery Research Laboratory, Department of Food and Drug Science, University of Parma, Parma, Italy; University Research Centre for the Innovation of Health Products, Biopharmanet-TEC, University of Parma, Parma, Italy.
| | - Ana Fortuna
- Laboratory of Pharmacology, Department of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT/ICNAS - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
57
|
Huang F, Baset A, Bello ST, Chen X, He J. Cholecystokinin facilitates the formation of long-term heterosynaptic plasticity in the distal subiculum. Commun Biol 2025; 8:153. [PMID: 39893259 PMCID: PMC11787286 DOI: 10.1038/s42003-025-07597-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 01/22/2025] [Indexed: 02/04/2025] Open
Abstract
It has been well established that Cornu Ammonis-(CA1) and subiculum (SUB) serve as the major output components of the entorhinal-hippocampal circuitry. Nevertheless, how the neuromodulators regulate the neurocircuitry in hippocampal formation has remained elusive. Cholecystokinin (CCK), is the most abundant neuropeptide in the central nervous system, which broadly regulates the animal's physiological status at multiple levels, including neuroplasticity and its behavioral consequences. Here, we uncover that exogenous CCK potentiates the excitatory synaptic transmission in the CA1-SUB projections via CCK-B receptor. Dual-color light theta burst stimulation elicits heterosynaptic long-term potentiation in distal SUB region. Light activation of medial entorhinal cortex (MEC) derived CCK-positive neurons triggers the CCK release in the SUB. Neuronal activities of SUB-projecting MECCCK neurons are necessary for conveying and processing of navigation-related information. In conclusion, our findings prove a crucial role of CCK in regulating neurobiological functions in the SUB region.
Collapse
Affiliation(s)
- Fengwen Huang
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong, China.
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, China.
| | - Abdul Baset
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, China
| | - Stephen Temitayo Bello
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, China
| | - Xi Chen
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong, China
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong, China
| | - Jufang He
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong, China.
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong, China.
| |
Collapse
|
58
|
Chevalier C, Tournier BB, Marizzoni M, Park R, Paquis A, Ceyzériat K, Badina AM, Lathuiliere A, Saleri S, Cillis FD, Cattaneo A, Millet P, Frisoni GB. Fecal Microbiota Transplantation (FMT) From a Human at Low Risk for Alzheimer's Disease Improves Short-Term Recognition Memory and Increases Neuroinflammation in a 3xTg AD Mouse Model. GENES, BRAIN, AND BEHAVIOR 2025; 24:e70012. [PMID: 39801363 PMCID: PMC11725982 DOI: 10.1111/gbb.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/15/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025]
Abstract
Human microbiota-associated murine models, using fecal microbiota transplantation (FMT) from human donors, help explore the microbiome's role in diseases like Alzheimer's disease (AD). This study examines how gut bacteria from donors with protective factors against AD influence behavior and brain pathology in an AD mouse model. Female 3xTgAD mice received weekly FMT for 2 months from (i) an 80-year-old AD patient (AD-FMT), (ii) a cognitively healthy 73-year-old with the protective APOEe2 allele (APOEe2-FMT), (iii) a 22-year-old healthy donor (Young-FMT), and (iv) untreated mice (Mice-FMT). Behavioral assessments included novel object recognition (NOR), Y-maze, open-field, and elevated plus maze tests; brain pathology (amyloid and tau), neuroinflammation (in situ autoradiography of the 18 kDa translocator protein in the hippocampus); and gut microbiota were analyzed. APOEe2-FMT improved short-term memory in the NOR test compared to AD-FMT, without significant changes in other behavioral tests. This was associated with increased neuroinflammation in the hippocampus, but no effect was detected on brain amyloidosis and tauopathy. Specific genera, such as Parabacteroides and Prevotellaceae_UGC001, were enriched in the APOEe2-FMT group and associated with neuroinflammation, while genera like Desulfovibrio were reduced and linked to decreased neuroinflammation. Gut microbiota from a donor with a protective factor against AD improved short-term memory and induced neuroinflammation in regions strategic to AD. The association of several genera with neuroinflammation in the APOEe2-FMT group suggests a collegial effect of the transplanted microbiome rather than a single-microbe driver effect. These data support an association between gut bacteria, glial cell activation, and cognitive function in AD.
Collapse
Affiliation(s)
- Claire Chevalier
- Département de Readaptation et gériatrieUniversity of GenevaGenevaSwitzerland
| | | | - Moira Marizzoni
- Biological Psychiatry UnitIRCCS Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - Rahel Park
- Département de Readaptation et gériatrieUniversity of GenevaGenevaSwitzerland
| | - Arthur Paquis
- Département de Readaptation et gériatrieUniversity of GenevaGenevaSwitzerland
| | - Kelly Ceyzériat
- Département de PsychiatrieUniversity of GenevaGenevaSwitzerland
| | | | | | - Samantha Saleri
- Biological Psychiatry UnitIRCCS Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - Floriana De Cillis
- Biological Psychiatry UnitIRCCS Centro San Giovanni di Dio FatebenefratelliBresciaItaly
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
| | - Annamaria Cattaneo
- Biological Psychiatry UnitIRCCS Centro San Giovanni di Dio FatebenefratelliBresciaItaly
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
| | - Philippe Millet
- Département de PsychiatrieUniversity of GenevaGenevaSwitzerland
| | - Giovanni B. Frisoni
- Département de Readaptation et gériatrieUniversity of GenevaGenevaSwitzerland
| |
Collapse
|
59
|
Borzage MT, Peterson BS. A Scoping Review of the Mechanisms Underlying Developmental Anesthetic Neurotoxicity. Anesth Analg 2025; 140:409-426. [PMID: 38536739 PMCID: PMC11427602 DOI: 10.1213/ane.0000000000006897] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2023] [Indexed: 09/28/2024]
Abstract
Although anesthesia makes painful or uncomfortable diagnostic and interventional health care procedures tolerable, it may also disrupt key cellular processes in neurons and glia, harm the developing brain, and thereby impair cognition and behavior in children. Many years of studies using in vitro, animal behavioral, retrospective database studies in humans, and several prospective clinical trials in humans have been invaluable in discerning the potential toxicity of anesthetics. The objective of this scoping review was to synthetize the evidence from preclinical studies for various mechanisms of toxicity across diverse experimental designs and relate their findings to those of recent clinical trials in real-world settings.
Collapse
Affiliation(s)
- Matthew Thomas Borzage
- From the Fetal and Neonatal Institute, Division of Neonatology, Children’s Hospital Los Angeles, Los Angeles, California
| | - Bradley S. Peterson
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California
- Institute for the Developing Mind, Children’s Hospital Los Angeles, Los Angeles, California
- Department of Psychiatry, Keck School of Medicine at the University of Southern California, Los Angeles, California
| |
Collapse
|
60
|
Gao Z, Gao Y, Li Y, Zhou J, Li G, Xie S, Jia R, Wang L, Jiang Z, Liang M, Du C, Chen Y, Liu Y, Du L, Wang C, Dou S, Lv Z, Wang L, Wang R, Shen B, Wang Z, Li Y, Han G. 5-HT 7R enhances neuroimmune resilience and alleviates meningitis by promoting CCR5 ubiquitination. J Adv Res 2025; 68:317-330. [PMID: 38432392 PMCID: PMC11785565 DOI: 10.1016/j.jare.2024.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/22/2024] [Accepted: 02/22/2024] [Indexed: 03/05/2024] Open
Abstract
INTRODUCTION Excessive immune activation induces tissue damage during infection. Compared to external strategies to reconstruct immune homeostasis, host balancing ways remain largely unclear. OBJECTIVES Here we found a neuroimmune way that prevents infection-induced tissue damage. METHODS By FACS and histopathology analysis of brain Streptococcus pneumonia meningitis infection model and behavioral testing. Western blot, co-immunoprecipitation, and ubiquitination analyze the Fluoxetine initiate 5-HT7R-STUB1-CCR5 K48-linked ubiquitination degradation. RESULTS Fluoxetine, a selective serotonin reuptake inhibitor, or the agonist of serotonin receptor 5-HT7R, protects mice from meningitis by inhibiting CCR5-mediated excessive immune response and tissue damage. Mechanistically, the Fluoxetine-5-HT7R axis induces proteasome-dependent degradation of CCR5 via mTOR signaling, and then recruits STUB1, an E3 ubiquitin ligase, to initiate K48-linked polyubiquitination of CCR5 at K138 and K322, promotes its proteasomal degradation. STUB1 deficiency blocks 5-HT7R-mediated CCR5 degradation. CONCLUSION Our results reveal a neuroimmune pathway that balances anti-infection immunity via happiness neurotransmitter receptor and suggest the 5-HT7R-CCR5 axis as a potential target to promote neuroimmune resilience.
Collapse
Affiliation(s)
- Zhenfang Gao
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Yang Gao
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Yuxiang Li
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Jie Zhou
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Ge Li
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Shun Xie
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Ruiyan Jia
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Lanying Wang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Ziying Jiang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Meng Liang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Chunxiao Du
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Yaqiong Chen
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Yinji Liu
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Lin Du
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Cong Wang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Shuaijie Dou
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Zhonglin Lv
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Lubin Wang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Renxi Wang
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Beifen Shen
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Zhiding Wang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China.
| | - Yunfeng Li
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China.
| | - Gencheng Han
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China.
| |
Collapse
|
61
|
Zhao JQ, Feng BY, Ye ZL, Ma XY, Du JZ, Li JM, Wu WL, Gao JJ, Li SJ, Peng SY, Huai JS, Ge LH, Lu CB. Activation of D2-like dopamine receptors improves the neuronal network and cognitive function of PPT1KI mice. Acta Pharmacol Sin 2025; 46:338-352. [PMID: 39284877 PMCID: PMC11747101 DOI: 10.1038/s41401-024-01377-7] [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: 01/25/2024] [Accepted: 08/08/2024] [Indexed: 01/22/2025]
Abstract
Palmitoyl-protein thioesterase 1 (PPT1) is a lysosomal depalmitoylation enzyme that mediates protein posttranslational modifications. Loss-of-function mutation of PPT1 causes a failure of the lysosomal degradation of palmitoylated proteins and results in a congenital disease characterized by progressive neuronal degeneration referred to as infantile neuronal ceroid lipofuscinosis (INCL). A mouse knock-in model of PPT1 (PPT1-KI) was established by introducing the R151X mutation into exon 5 of the PPT1 gene, which exhibited INCL-like pathological lesions. We previously reported that hippocampal γ oscillations were impaired in PPT1 mice. Hippocampal γ oscillations can be enhanced by selective activation of the dopamine D4 receptor (DR4), a dopamine D2-like receptor. In this study, we investigated the changes in DR expression and the effects of dopamine and various DR agonists on neural network activity, cognition and motor function in PPT1KI mice. Cognition and motor defects were evaluated via Y-maze, novel object recognition and rotarod tests. Extracellular field potentials were elicited in hippocampal slices, and neuronal network oscillations in the gamma frequency band (γ oscillations) were induced by perfusion with kainic acid (200 nM). PPT1KI mice displayed progressive impairments in γ oscillations and hippocampus-related memory, as well as abnormal expression profiles of dopamine receptors with preserved expression of DR1 and 3, increased membrane expression of DR4 and decreased DR2 levels. The immunocytochemistry analysis revealed the colocalization of PPT1 with DR4 or DR2 in the soma and large dendrites of both WT and PPT1KI mice. Immunoprecipitation confirmed the interaction between PPT1 and DR4 or DR2. The impaired γ oscillations and cognitive functions were largely restored by the application of exogenous dopamine, the selective DR2 agonist quinpirole or the DR4 agonist A412997. Furthermore, the administration of A412997 (0.5 mg/kg, i.p.) significantly upregulated the activity of CaMKII in the hippocampus of 5-month-old PPT1KI mice. Collectively, these results suggest that the activation of D2-like dopamine receptors improves cognition and network activity in PPT1KI mice and that specific DR subunits may be potential targets for the intervention of neurodegenerative disorders, such as INCL.
Collapse
Affiliation(s)
- Jun-Qiang Zhao
- First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 435000, China
- School of Medical Imaging, Xinxiang Medical University, Xinxiang, 435000, China
| | - Bing-Yan Feng
- First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 435000, China
| | - Zhen-Li Ye
- Henan International Joint Laboratory for Noninvasive Neuromodulation/Key Laboratory of Brain Research of Henan Province, Department of Physiology & Pathophysiology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, 435000, China
| | - Xiao-Yin Ma
- Henan International Joint Laboratory for Noninvasive Neuromodulation/Key Laboratory of Brain Research of Henan Province, Department of Physiology & Pathophysiology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, 435000, China
| | - Jing-Zhi Du
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 435000, China
| | - Jun-Mei Li
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 435000, China
| | - Wan-Liu Wu
- First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 435000, China
| | - Jing-Jing Gao
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 435000, China
| | - Song-Ji Li
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 435000, China
| | - Shi-Yong Peng
- Henan International Joint Laboratory for Noninvasive Neuromodulation/Key Laboratory of Brain Research of Henan Province, Department of Physiology & Pathophysiology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, 435000, China
| | - Ji-Sen Huai
- Henan International Joint Laboratory for Noninvasive Neuromodulation/Key Laboratory of Brain Research of Henan Province, Department of Physiology & Pathophysiology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, 435000, China
| | - Li-Hao Ge
- Henan International Joint Laboratory for Noninvasive Neuromodulation/Key Laboratory of Brain Research of Henan Province, Department of Physiology & Pathophysiology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, 435000, China.
| | - Cheng-Biao Lu
- First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 435000, China.
- Henan International Joint Laboratory for Noninvasive Neuromodulation/Key Laboratory of Brain Research of Henan Province, Department of Physiology & Pathophysiology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, 435000, China.
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 435000, China.
| |
Collapse
|
62
|
Dutton AJ, Turnbaugh EM, Patel CD, Garland CR, Taylor SA, Alers-Velazquez R, Knipe DM, Nautiyal KM, Leib DA. Asymptomatic neonatal herpes simplex virus infection in mice leads to persistent CNS infection and long-term cognitive impairment. PLoS Pathog 2025; 21:e1012935. [PMID: 39919123 PMCID: PMC11828378 DOI: 10.1371/journal.ppat.1012935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/14/2025] [Accepted: 01/25/2025] [Indexed: 02/09/2025] Open
Abstract
Neonatal herpes simplex virus (nHSV) is a devastating infection impacting approximately 14,000 newborns globally each year. nHSV infection is associated with high neurologic morbidity and mortality, making early intervention critical. Clinical outcomes of symptomatic nHSV infections are well-studied, but little is known about the frequency of, or outcomes following, subclinical or asymptomatic nHSV. Given the ubiquitous nature of HSV infection and frequency of asymptomatic shedding in adults, subclinical infections are underreported and could contribute to long-term neurological damage. To assess potential neurological morbidity associated with subclinical nHSV infection, we developed a low-dose (100 PFU) intranasal HSV infection model in neonatal wild-type C57BL/6 mice. At this dose, HSV DNA was detected in the brain by quantitative PCR (qPCR) but was not associated with acute clinical signs of infection. However, months after neonatal inoculation with this low dose of HSV, we observed impaired mouse performance on a range of cognitive and memory tests. Memory impairment was induced by infection with either HSV-1 or HSV-2 wild-type viruses, indicating that the cognitive impairment associated with neonatal infection was not strain-specific. Maternal immunization reduced neonate central nervous system (CNS) viral burden and prevented offspring from developing neurological sequelae following nHSV infection. Altogether, these results support the idea that subclinical neonatal infections may lead to cognitive decline in adulthood and that maternal vaccination is an effective strategy for reducing neurological sequelae in infected offspring. These findings may have profound implications for understanding and modeling the etiology of human neurodegenerative disorders such as Alzheimer's Disease.
Collapse
Affiliation(s)
- Abigail J. Dutton
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
- Guarini School of Graduate and Advanced Studies at Dartmouth, Hanover, New Hampshire, United States of America
| | - Evelyn M. Turnbaugh
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
- Guarini School of Graduate and Advanced Studies at Dartmouth, Hanover, New Hampshire, United States of America
| | - Chaya D. Patel
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
- Guarini School of Graduate and Advanced Studies at Dartmouth, Hanover, New Hampshire, United States of America
| | - Callaghan R. Garland
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| | - Sean A. Taylor
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| | - Roberto Alers-Velazquez
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| | - David M. Knipe
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Katherine M. Nautiyal
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, New Hampshire, United States of America
| | - David A. Leib
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| |
Collapse
|
63
|
Tang X, Zhu C, Liu TC, Zhu R, Deng G, Zhou P, Liu D. Sunflower Oil Fortified with Vitamins D and A and Sunflower Lecithin Ameliorated Scopolamine-Induced Cognitive Dysfunction in Mice and Exploration of the Underlying Protective Pathways. Nutrients 2025; 17:553. [PMID: 39940410 PMCID: PMC11819866 DOI: 10.3390/nu17030553] [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: 01/06/2025] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
The incidence of cognitive disorders is increasing globally, with a reported prevalence of over 50 million individuals affected, and current interventions offer limited efficacy. This study investigates the effects of sunflower oil fortified with sunflower lecithin, vitamin D, and vitamin A on scopolamine-induced cognitive dysfunction in mice and explores the underlying mechanisms. The incidence of cognitive disorders, such as Alzheimer's disease, is increasing yearly, and current interventions offer limited efficacy. Therefore, this research aims to evaluate the cognitive improvement effects of the three added functional factors on mice with learning and memory impairments, along with the associated molecular mechanisms. Behavioral tests, biochemical assays, and real-time quantitative polymerase chain reaction (RT-qPCR) were utilized to examine the intervention effects of these functional factors on scopolamine-induced cognitive impairment in mice. The results revealed that the groups treated with sunflower lecithin and vitamin D significantly enhanced the mice's exploratory behavior, working memory, and spatial memory, with increases of 1.6 times and 4.5 times, respectively, in the open field and novel object recognition tests (VD group). Additionally, these treatments reduced levels of inflammatory markers and IL-6, increased antioxidant GSH levels, and decreased oxidative stress marker MDA levels, with all effects showing significant differences (p < 0.01). The effects were further enhanced when vitamin A was combined with these treatments. Transcriptomic analysis demonstrated that the intervention groups had markedly improved learning and memory abilities through upregulation of key gene expression levels in the PI3K-AKT signaling pathway, cholinergic pathway, and folate biosynthesis pathway. These findings provide a theoretical basis for the development of nutritionally fortified edible oils with added sunflower lecithin, vitamin D, and vitamin A, which may help prevent and ameliorate cognitive disorders.
Collapse
Grants
- Standard Foods (China) Co., Ltd., No. 88 Dalian West Road, Taicang Port Economic and Technological Development Zone New Zone, Suzhou, Jiangsu 215400, China Standard Foods (China) Co., Ltd., No. 88 Dalian West Road, Taicang Port Economic and Technological Development Zone New Zone, Suzhou, Jiangsu 215400, China
Collapse
Affiliation(s)
- Xue Tang
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (X.T.); (C.Z.); (R.Z.); (G.D.)
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China; (P.Z.); (D.L.)
| | - Chengkai Zhu
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (X.T.); (C.Z.); (R.Z.); (G.D.)
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China; (P.Z.); (D.L.)
| | - Tristan C. Liu
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (X.T.); (C.Z.); (R.Z.); (G.D.)
| | - Rongxiang Zhu
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (X.T.); (C.Z.); (R.Z.); (G.D.)
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China; (P.Z.); (D.L.)
| | - Guoliang Deng
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (X.T.); (C.Z.); (R.Z.); (G.D.)
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China; (P.Z.); (D.L.)
| | - Peng Zhou
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China; (P.Z.); (D.L.)
| | - Dasong Liu
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China; (P.Z.); (D.L.)
| |
Collapse
|
64
|
O’Neill E, Curham L, Ní Chasaide C, O’Brien S, McManus G, Moran B, Rubin K, Glazer S, Lynch MA, Mills KH. Neonatal infection with Bordetella pertussis promotes autism-like phenotypes in mice. iScience 2025; 28:111548. [PMID: 39897939 PMCID: PMC11784780 DOI: 10.1016/j.isci.2024.111548] [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: 04/11/2024] [Revised: 09/09/2024] [Accepted: 12/04/2024] [Indexed: 02/04/2025] Open
Abstract
Autism spectrum disorder (ASD) has been linked with infections early in life. Here we demonstrate that the infection of neonatal mice with the respiratory pathogen Bordetella pertussis leads to neuroinflammation, neurodevelopmental defects, and ASD-like behaviors. Following the respiratory challenge of neonatal mice with B. pertussis, multiple atypical CNS findings were observed, including blood-brain barrier disruption, dissemination of live B. pertussis bacteria to the brain with the concomitant infiltration of inflammatory monocytes, neutrophils, and activated IL-17A- and IFN-γ-producing CD4 T cells. Microglia from infected mice were activated, with impaired phagocytic function, resulting in defective synaptic pruning and disrupted neuronal circuit formation. Impaired neurodevelopment in B. pertussis-infected post-natal mice was associated with ASD-like behavioral abnormalities in young adulthood. Our data indicate that infection with virulent B. pertussis during infancy increases the risk of autism-like behavior in young adult mice. A study into the potential role of B. pertussis in human ASD is warranted.
Collapse
Affiliation(s)
- Eoin O’Neill
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, D02PD91 Dublin, Ireland
| | - Lucy Curham
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| | - Caitlín Ní Chasaide
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| | - Síofra O’Brien
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| | - Gavin McManus
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| | - Barry Moran
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| | - Keith Rubin
- ILiAD Biotechnologies, Weston, FL 33331, USA
| | | | - Marina A. Lynch
- Trinity College Institute of Neuroscience, Trinity College Dublin, D02PD91 Dublin, Ireland
| | - Kingston H.G. Mills
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| |
Collapse
|
65
|
Hoisington ZW, Gangal H, Phamluong K, Shukla C, Ehinger Y, Moffat JJ, Homanics GE, Wang J, Ron D. Prosapip1 in the dorsal hippocampus mediates synaptic protein composition, long-term potentiation, and spatial memory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.06.13.597459. [PMID: 38915579 PMCID: PMC11195216 DOI: 10.1101/2024.06.13.597459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Prosapip1 is a brain-specific protein localized to the postsynaptic density, where it promotes dendritic spine maturation in primary hippocampal neurons. However, nothing is known about the role of Prosapip1 in vivo. To examine this, we utilized the Cre-loxP system to develop a Prosapip1 neuronal knockout mouse. We found that Prosapip1 controls the synaptic localization of its binding partner SPAR, along with PSD-95 and the GluN2B subunit of the NMDA receptor (NMDAR) in the dorsal hippocampus (dHP). We next sought to identify the potential contribution of Prosapip1 to the activity and function of the NMDAR and found that Prosapip1 plays an important role in NMDAR-mediated transmission and long-term potentiation (LTP) in the CA1 region of the dHP. As LTP is the cellular hallmark of learning and memory, we examined the consequences of neuronal knockout of Prosapip1 on dHP-dependent memory. We found that global or dHP-specific neuronal knockout of Prosapip1 caused a deficit in learning and memory whereas developmental, locomotor, and anxiety phenotypes were normal. Taken together, Prosapip1 in the dHP promotes the proper localization of synaptic proteins which, in turn, facilitates LTP driving recognition, social, and spatial learning and memory.
Collapse
|
66
|
Duarte RMF, Ribeiro-Barbosa ER, Ferreira FR, Espindola FS, Spini VBMG. Resveratrol prevents offspring's behavioral impairment associated with immunogenic stress during pregnancy. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111188. [PMID: 39522792 DOI: 10.1016/j.pnpbp.2024.111188] [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: 02/02/2024] [Revised: 10/31/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
Evidence suggests that prenatal maternal immunological stress is associated with an increased risk of neurological and psychiatric disorders in the developing offspring. Protecting the embryo during this critical period of neurodevelopment, when the brain is especially vulnerable, is therefore crucial. Polyphenols, with their antioxidant and anti-inflammatory properties, offer promising therapeutic approaches. This study demonstrated a series of behavioral changes induced by maternal immune activation (MIA) triggered by an antigenic solution derived from the H1N1 virus. These changes include significant differences in anxiety and risk assessment behaviors, increased immobility in the forced swim test, impairments in memory and object recognition, and social deficits resembling autism. The phenolic compound resveratrol (RSV) was evaluated for its in vitro antioxidant capacity and characterized using infrared spectroscopy. Administering RSV from embryonic day 14 (E14) to embrionyc day 19 (E19) during MIA effectively reduced its harmful effects on the offspring. This was evidenced by a significant restoration of social behaviors, memory, and recognition, as well as anxiolytic and antidepressant effects in the adult offspring. These findings contribute to new therapeutic strategies for preventing psychiatric disorders associated with neurodevelopmental stressors.
Collapse
Affiliation(s)
- Rener Mateus Francisco Duarte
- Department of Biochemistry and Molecular Biology, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia, Brazil.
| | - Erika Renata Ribeiro-Barbosa
- Department of Physiological Sciences, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | | | - Foued Salmen Espindola
- Department of Biochemistry and Molecular Biology, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia, Brazil
| | | |
Collapse
|
67
|
Sulimai N, Brown J, Lominadze D. Caffeic Acid Phenethyl Ester Protects Neurons Against Oxidative Stress and Neurodegeneration During Traumatic Brain Injury. Biomolecules 2025; 15:80. [PMID: 39858474 PMCID: PMC11762460 DOI: 10.3390/biom15010080] [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: 12/10/2024] [Revised: 01/04/2025] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Traumatic brain injury (TBI) is an inflammatory disease causing neurodegeneration. One of the consequences of inflammation is an elevated blood level of fibrinogen (Fg). Earlier we found that extravasated Fg induced an increased expression of neuronal nuclear factor kappa B (NF-κB) p65. In the present study, we aimed to evaluate the effect of caffeic acid phenethyl ester (CAPE), an inhibitor of NF-κB, on Fg-induced neurodegeneration in vitro and in mice with mild-to-moderate TBI. Primary mouse brain cortical neurons were treated with Fg (0.5 or 1 mg/mL) in the presence or absence of CAPE. A cortical contusion injury -induced model of TBI in C57BL/6 mice was used. Mice were treated with CAPE for two weeks. The generation of reactive oxygen species (ROS) and neuronal viability were assessed. Mice memory was assessed using novel object recognition and contextual fear conditioning tests. The generation of ROS and viability of neurons in vitro and in the brain samples were assessed. Data showed that CAPE attenuated the Fg-induced generation of ROS and neuronal death. CAPE improved the cognitive function of the mice with TBI. The results suggest that Fg-induced generation of ROS could be a mechanism involved in cognitive impairment and that CAPE can offer protection against oxidative damage and neurodegeneration.
Collapse
Affiliation(s)
- Nurul Sulimai
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (N.S.); (J.B.)
| | - Jason Brown
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (N.S.); (J.B.)
| | - David Lominadze
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (N.S.); (J.B.)
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| |
Collapse
|
68
|
Aulston BD, Gimse K, Bazick HO, Kramar EA, Pizzo DP, Parra-Rivas LA, Sun J, Branes-Guerrero K, Checka N, Bagheri N, Satyadev N, Carlson-Stevermer J, Saito T, Saido TC, Audhya A, Wood MA, Zylka MJ, Saha K, Roy S. Long term rescue of Alzheimer's deficits in vivo by one-time gene-editing of App C-terminus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.06.08.598099. [PMID: 38895278 PMCID: PMC11185791 DOI: 10.1101/2024.06.08.598099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Gene-editing technologies promise to create a new class of therapeutics that can achieve permanent correction with a single intervention. Besides eliminating mutant alleles in familial disease, gene-editing can also be used to favorably manipulate upstream pathophysiologic events and alter disease-course in wider patient populations, but few such feasible therapeutic avenues have been reported. Here we use CRISPR-Cas9 to edit the last exon of amyloid precursor protein (App), relevant for Alzheimer's disease (AD). Our strategy effectively eliminates an endocytic (YENPTY) motif at APP C-terminus, while preserving the N-terminus and compensatory APP-homologues. This manipulation favorably alters events along the amyloid-pathway - inhibiting toxic APP-β-cleavage fragments (including Aβ) and upregulating neuroprotective APP-α-cleavage products. AAV-driven editing ameliorates neuropathologic, electrophysiologic, and behavioral deficits in an AD knockin mouse model. Effects persist for many months, and no abnormalities are seen in WT mice even after germline App-editing; underlining overall efficacy and safety. Pathologic alterations in the glial-transcriptome of App-KI mice, as seen by single nuclei RNA-sequencing (sNuc-Seq), are also normalized by App C-terminus editing. Our strategy takes advantage of innate transcriptional rules that render terminal exons insensitive to nonsense-decay, and the upstream manipulation is expected to be effective for all forms of AD. These studies offer a path for a one-time disease-modifying treatment for AD.
Collapse
Affiliation(s)
- Brent D Aulston
- Department of Pathology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, USA
| | - Kirstan Gimse
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Hannah O Bazick
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Eniko A Kramar
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, USA
| | - Donald P Pizzo
- Department of Pathology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, USA
| | - Leonardo A Parra-Rivas
- Department of Pathology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, USA
| | - Jichao Sun
- Department of Pathology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, USA
- Present address: Department of Geriatrics and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Kristen Branes-Guerrero
- Department of Pathology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, USA
- Present address: Eli Lilly Pharmaceuticals, Indianapolis, IN, USA
| | - Nidhi Checka
- Department of Pathology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, USA
| | - Neda Bagheri
- Department of Pathology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, USA
| | - Nihal Satyadev
- Department of Pathology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, USA
- Present address: Department of Neurology, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Jared Carlson-Stevermer
- Synthego Corporation, 3696 Haven Ave Suite A, Redwood City, CA 94063
- Present address: Serotiny Inc., 329 Oyster Point Boulevard, 3rd Floor, South San Francisco, CA 94080
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Anjon Audhya
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Marcelo A Wood
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, USA
| | - Mark J Zylka
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Krishanu Saha
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Subhojit Roy
- Department of Pathology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, USA
- Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, USA
| |
Collapse
|
69
|
Masson BA, Kiridena P, Lu D, Kleeman EA, Reisinger SN, Qin W, Davies WJ, Muralitharan RR, Jama HA, Antonacci S, Marques FZ, Gubert C, Hannan AJ. Depletion of the paternal gut microbiome alters sperm small RNAs and impacts offspring physiology and behavior in mice. Brain Behav Immun 2025; 123:290-305. [PMID: 39293692 DOI: 10.1016/j.bbi.2024.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024] Open
Abstract
The paternal environment prior to conception has been demonstrated to influence offspring physiology and behavior, with the sperm epigenome (including noncoding RNAs) proposed as a potential facilitator of non-genetic inheritance. Whilst the maternal gut microbiome has been established as an important influence on offspring development, the impact of the paternal gut microbiome on offspring development, health and behavior is largely unknown. Gut microbiota have major influences on immunity, and thus we hypothesized that they may be relevant to paternal immune activation (PIA) modulating epigenetic inheritance in mice. Therefore, male C57BL/6J mice (F0) were orally administered non-absorbable antibiotics via drinking water in order to substantially deplete their gut microbiome. Four weeks after administration of the antibiotics (gut microbiome depletion), F0 male mice were then mated with naïve female mice. The F1 offspring of the microbiome-depleted males had reduced body weight as well as altered gut morphology (shortened colon length). F1 females showed significant alterations in affective behaviors, including measures of anxiety and depressive-like behaviors, indicating altered development. Analysis of small noncoding RNAs in the sperm of F0 mice revealed that gut microbiome depletion is associated with differential expression of 8 different PIWI-interacting RNAs (piRNAs), each of which has the potential to modulate the expression of multiple downstream gene targets, and thus influence epigenetic inheritance and offspring development. This study demonstrates that the gut-germline axis influences sperm small RNA profiles and offspring physiology, with specific impacts on offspring affective and/or coping behaviors. These findings may have broader implications for other animal species with comparable gut microbiota, intergenerational epigenetics and developmental biology, including humans.
Collapse
Affiliation(s)
- Bethany A Masson
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Pamudika Kiridena
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Da Lu
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Elizabeth A Kleeman
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Sonali N Reisinger
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Wendy Qin
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - William J Davies
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Rikeish R Muralitharan
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia; Victorian Heart Institute, Monash University, Clayton, Australia
| | - Hamdi A Jama
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia
| | - Simona Antonacci
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia; Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Victorian Heart Institute, Monash University, Clayton, Australia
| | - Carolina Gubert
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia.
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia; Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
70
|
JianHua Z, Li M, Hu Q, Donoghue P, Jiang S, Li J, Li S, Ren X, Zhang Z, Du J, Yu Y, Chazot P, Lu C. CaMKIIα-TARPγ8 signaling mediates hippocampal synaptic impairment in aging. Aging Cell 2025; 24:e14349. [PMID: 39380368 PMCID: PMC11709088 DOI: 10.1111/acel.14349] [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: 03/14/2024] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 10/10/2024] Open
Abstract
Aging-related decline in memory and synaptic function are associated with the dysregulation of calcium homeostasis, attributed to the overexpression of voltage-gated calcium channels (VGCC). The membrane insertion of AMPAR governed by the AMPAR auxiliary proteins is essential for synaptic transmission and plasticity (LTP). In this study, we demonstrated the hippocampal expression of the transmembrane AMPAR regulatory proteins γ-8 (TARPγ8) was reduced in aged mice along with the reduced CaMKIIα activity and memory impairment. We further showed that TARPγ8 expression was dependent on CaMKIIα activity. Inhibition of CaMKIIα activity significantly reduced the hippocampal TARPγ8 expression and CA3-CA1 LTP in young mice to a similar level to that of the aged mice. Furthermore, the knockdown of hippocampal TARPγ8 impaired LTP and memory in young mice, which mimicked the aging-related changes. We confirmed the enhanced hippocampal VGCC (Cav-1.3) expression in aged mice and found that inhibition of VGCC activity largely increased both p-CaMKIIα and TARPγ8 expression in aged mice, whereas inhibition of NMDAR or Calpains had no effect. In addition, we found that the exogenous expression of human TARPγ8 in the hippocampus in aged mice restored LTP and memory function. Collectively, these results indicate that the synaptic and cognitive impairment in aging is associated with the downregulation of CaMKIIα-TARPγ8 signaling caused by VGCC activation. Our results suggest that TARPγ8 may be a key molecular biomarker for brain aging and that boosting CaMKIIα-TARPγ8 signaling may be critical for the restoration of synaptic plasticity of aging and aging-related diseases.
Collapse
Affiliation(s)
- Zhao JianHua
- Henan International Joint Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of NeurologyFirst Affiliated Hospital of Xinxiang Medical UniversityWeihui, XinxiangHenanChina
- Henan International Joint Laboratory of Non‐Invasive Neuromodulation, Department of Physiology and PathophysiologyXinxiang Medical UniversityXinxiangHenanChina
| | - MingCan Li
- Henan International Joint Laboratory of Non‐Invasive Neuromodulation, Department of Physiology and PathophysiologyXinxiang Medical UniversityXinxiangHenanChina
- Institute of Psychiatry and Neuroscience, Xinxiang Medical UniversityXinxiangHenanChina
| | - Qilin Hu
- Henan International Joint Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of NeurologyFirst Affiliated Hospital of Xinxiang Medical UniversityWeihui, XinxiangHenanChina
- School of Medical EngineeringXinxiang Medical UniversityXinxiangChina
| | - Peter Donoghue
- Department of BiosciencesWolfson Research Institute for Health and Wellbeing, Durham UniversityDurhamUK
| | - Sanwei Jiang
- Henan International Joint Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of NeurologyFirst Affiliated Hospital of Xinxiang Medical UniversityWeihui, XinxiangHenanChina
| | - Junmei Li
- Henan International Joint Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of NeurologyFirst Affiliated Hospital of Xinxiang Medical UniversityWeihui, XinxiangHenanChina
- Institute of Psychiatry and Neuroscience, Xinxiang Medical UniversityXinxiangHenanChina
| | - Songji Li
- Henan International Joint Laboratory of Non‐Invasive Neuromodulation, Department of Physiology and PathophysiologyXinxiang Medical UniversityXinxiangHenanChina
| | - Xinyi Ren
- Henan International Joint Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of NeurologyFirst Affiliated Hospital of Xinxiang Medical UniversityWeihui, XinxiangHenanChina
| | - Ziyuan Zhang
- Henan International Joint Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of NeurologyFirst Affiliated Hospital of Xinxiang Medical UniversityWeihui, XinxiangHenanChina
| | - Jingzhi Du
- Henan International Joint Laboratory of Non‐Invasive Neuromodulation, Department of Physiology and PathophysiologyXinxiang Medical UniversityXinxiangHenanChina
| | - Yi Yu
- School of Medical EngineeringXinxiang Medical UniversityXinxiangChina
| | - Paul Chazot
- Department of BiosciencesWolfson Research Institute for Health and Wellbeing, Durham UniversityDurhamUK
| | - Chengbiao Lu
- Henan International Joint Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of NeurologyFirst Affiliated Hospital of Xinxiang Medical UniversityWeihui, XinxiangHenanChina
- Henan International Joint Laboratory of Non‐Invasive Neuromodulation, Department of Physiology and PathophysiologyXinxiang Medical UniversityXinxiangHenanChina
- Institute of Psychiatry and Neuroscience, Xinxiang Medical UniversityXinxiangHenanChina
| |
Collapse
|
71
|
Zhang Y, Chen J, Li Y, Jiao B, Luo S. Disease-modifying therapies for Alzheimer's disease: Clinical trial progress and opportunity. Ageing Res Rev 2025; 103:102595. [PMID: 39581354 DOI: 10.1016/j.arr.2024.102595] [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: 10/24/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024]
Abstract
The U.S. Food and Drug Administration (FDA) recently approved lecanemab and donanemab for the treatment of early symptomatic Alzheimer's disease (AD) after their phase III trials reached endpoints. These two anti-amyloid β monoclonal antibodies represent the latest promise of disease-modifying therapy (DMT) for AD, which undoubtedly reignites new hope for DMTs to combat the staggering financial and human costs of AD. However, in addition to these two successful antibodies, there have been enormous efforts to develop DMTs in various aspects to meet the therapeutic requirement of AD. In this review, we delineate the core principles and methodologies of diverse DMTs, covering the advances in clinical trials of drug candidates that either have been discontinued, completed, or are ongoing, as well as brain stimulation and lifestyle interventions. In addition, by overseeing the fate of various candidate molecules, we hope to provide references and ideas for prospective approaches and promising applications of DTMs for AD, particularly in terms of universality and clinical application economics, to optimize efficacy and maximize AD patient benefits in the future.
Collapse
Affiliation(s)
- Yujie Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Xiangya School of Medicine, Central South University, Changsha 410013, PR China
| | - Jie Chen
- Department of Rehabilitation, Xiangya Boai Rehabilitation Hospital, Changsha 410100, PR China
| | - Yanru Li
- Department of Rehabilitation, Xiangya Boai Rehabilitation Hospital, Changsha 410100, PR China
| | - Bin Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, PR China; National Clinical Research Center for Geriatric Disorders, Central South University, Changsha 410008, PR China; Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha 410000, PR China; Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha 410008, PR China
| | - Shilin Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, PR China; National Clinical Research Center for Geriatric Disorders, Central South University, Changsha 410008, PR China; Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha 410000, PR China; Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha 410008, PR China.
| |
Collapse
|
72
|
Lin S, Zhu G, Xie J, Wen X, Deng L, Li S, Liu G, Wang F, Chen S, Xiao Z. Study on the Therapeutic Effects of Bisdemethoxycurcumin on a Cerebral Amyloid Angiopathy Mouse Model Established via Chronic Treatment With Five Vascular Risk Factors. Brain Behav 2025; 15:e70245. [PMID: 39829173 PMCID: PMC11743980 DOI: 10.1002/brb3.70245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/12/2024] [Accepted: 12/14/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND AND PURPOSE Cerebral amyloid angiopathy (CAA) is recognized as a major contributor to progressive cognitive decline and cerebral hemorrhages in the elderly population. Currently, there is a global shortage of safe and effective treatments for this condition. Bisdemethoxycurcumin (BDMC) has been demonstrated to exhibit pharmacological effects with anti-Aβ toxicity properties. Thus, the present study mainly focused on the potential therapeutic effects of BDMC on CAA. METHOD The 30 male C57BL/6 mice were subjected to chronic treatment with five vascular risk factors (lipopolysaccharide, social stress, streptozotocin, high-cholesterol diet, and copper-containing drinking water) for 35 weeks to establish a CAA mouse model. Of these, 15 CAA mice received oral administration of BDMC (50 mg/kg) for two consecutive weeks as an intervention, while the remaining 15 CAA mice received an equal volume of physiological saline by gavage. The study observed the levels of Aβ40 and proinflammatory factors in brain tissue and plasma, Aβ deposition in cerebral blood vessels, microbleeds in brain tissue, expression of proteins related to the cGAS/STING signaling pathway in brain tissue, as well as the contents of p-RIPK-1, p-RIPK-3, p-MLKL, neuronal morphology, and learning and memory abilities in mice. RESULT The therapeutic administration of BDMC demonstrates a pronounced efficacy in alleviating Aβ burden and cerebral microbleeding in CAA mice, concurrently enhancing learning and memory capabilities. Interestingly, BDMC may inhibits neuroinflammatory responses by reducing the expression of cGAS/STING signaling pathway proteins and suppresses necroptosis. CONCLUSION Our research findings demonstrate that BDMC exerts therapeutic effects in a mouse model of CAA established through chronic treatment involving five vascular risk factors.
Collapse
Affiliation(s)
- Shudong Lin
- Department of Neurology, The First Affiliated Hospital, Multi‐Omics Research Center for Brain Disorders, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
- Clinical Research Center for Immune‐Related Encephalopathy In Hunan Province, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
| | - Guanghua Zhu
- Department of Neurology, The First Affiliated Hospital, Multi‐Omics Research Center for Brain Disorders, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
- Clinical Research Center for Immune‐Related Encephalopathy In Hunan Province, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
| | - Juan Xie
- Department of Neurology, The First Affiliated Hospital, Multi‐Omics Research Center for Brain Disorders, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
- Department of Emergency, The First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunanPeople's Republic of China
| | - Xuanwei Wen
- Department of Neurology, The First Affiliated Hospital, Multi‐Omics Research Center for Brain Disorders, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
- Clinical Research Center for Immune‐Related Encephalopathy In Hunan Province, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
| | - Limin Deng
- Department of Neurology, The First Affiliated Hospital, Multi‐Omics Research Center for Brain Disorders, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
- Clinical Research Center for Immune‐Related Encephalopathy In Hunan Province, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
| | - Sijing Li
- Department of Neurology, The First Affiliated Hospital, Multi‐Omics Research Center for Brain Disorders, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
- Clinical Research Center for Immune‐Related Encephalopathy In Hunan Province, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
| | - Guozhi Liu
- Department of Neurology, The First Affiliated Hospital, Multi‐Omics Research Center for Brain Disorders, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
- Clinical Research Center for Immune‐Related Encephalopathy In Hunan Province, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
| | - Feiyan Wang
- Department of Neurology, The First Affiliated Hospital, Multi‐Omics Research Center for Brain Disorders, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
- Clinical Research Center for Immune‐Related Encephalopathy In Hunan Province, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
| | - Shuangxi Chen
- Department of Neurology, The First Affiliated Hospital, Multi‐Omics Research Center for Brain Disorders, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
- Clinical Research Center for Immune‐Related Encephalopathy In Hunan Province, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
| | - Zijian Xiao
- Department of Neurology, The First Affiliated Hospital, Multi‐Omics Research Center for Brain Disorders, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
- Clinical Research Center for Immune‐Related Encephalopathy In Hunan Province, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
| |
Collapse
|
73
|
Brandão‐Teles C, Carregari VC, Reis‐de‐Oliveira G, Smith BJ, Chaves Y, Sousa Santos AV, Pinheiro EMDC, Oliveira CC, Vieira AS, Crunfli F, Martins‐de‐Souza D. Impacts of hnRNP A1 Splicing Inhibition on the Brain Remyelination Proteome. J Neurochem 2025; 169:e16304. [PMID: 39840781 PMCID: PMC11752419 DOI: 10.1111/jnc.16304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/26/2024] [Accepted: 12/16/2024] [Indexed: 01/23/2025]
Abstract
Oligodendrocytes, the myelinating cells in the central nervous system, are implicated in several neurological disorders marked by dysfunctional RNA-binding proteins (RBPs). The present study aimed at investigating the role of hnRNP A1 in the proteome of the corpus callosum, prefrontal cortex, and hippocampus of a murine cuprizone-induced demyelination model. Right after the cuprizone insult, we administered an hnRNP A1 splicing activity inhibitor and analyzed its impact on brain remyelination by nanoESI-LC-MS/MS label-free proteomic analysis to assess the biological processes affected in these brain regions. Significant alterations in essential myelination proteins highlighted the involvement of hnRNP A1 in maintaining myelin integrity. Pathways related to sphingolipid and endocannabinoid signaling were affected, as well as the synaptic vesicle cycle and GABAergic synapses. Although behavioral impairments were not observed, molecular changes suggest potential links to memory, synaptic function, and neurotransmission processes. These findings enhance our understanding of the multifaceted roles of hnRNP A1 in the central nervous system, providing valuable insights for future investigations and therapeutic interventions in neurodegenerative and demyelinating diseases.
Collapse
Affiliation(s)
- Caroline Brandão‐Teles
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of BiologyUniversity of CampinasCampinasBrazil
| | - Victor Corasolla Carregari
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of BiologyUniversity of CampinasCampinasBrazil
| | - Guilherme Reis‐de‐Oliveira
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of BiologyUniversity of CampinasCampinasBrazil
| | - Bradley J. Smith
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of BiologyUniversity of CampinasCampinasBrazil
| | - Yane Chaves
- Translational Neuropsychiatry Unit, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Aline Valéria Sousa Santos
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of BiologyUniversity of CampinasCampinasBrazil
| | | | - Caio C. Oliveira
- Institute of ChemistryUniversity of CampinasCampinasSão PauloBrazil
| | - Andre Schwambach Vieira
- Laboratory of Electrophysiology, Neurobiology and Behaviour, Department of Functional and Structural Biology, Institute of BiologyUniversity of CampinasCampinasBrazil
| | - Fernanda Crunfli
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of BiologyUniversity of CampinasCampinasBrazil
- Translational Neuropsychiatry Unit, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Daniel Martins‐de‐Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of BiologyUniversity of CampinasCampinasBrazil
- Experimental Medicine Research Cluster (EMRC)University of CampinasCampinasSPBrazil
- D'Or Institute for Research and Education (IDOR)São PauloBrazil
- Instituto Nacional de Biomarcadores Em Neuropsiquiatria, Conselho Nacional de Desenvolvimento Científico e TecnológicoSão PauloBrazil
- INCT in Modelling Human Complex Diseases With 3D Platforms (Model3D), Conselho Nacional de Desenvolvimento Científico e TecnológicoSão PauloBrazil
| |
Collapse
|
74
|
Chen TY, Chen YR, Hsu ML, Liao YT, Wu CH, Yao CA, Yang WC, Lin W, Lin Y. Homoplantaginin Antagonizes N-Methyl-d-aspartate Receptor and Extracellular Signal-Regulated Kinase Signaling in Aβ Oligomers-Induced Neuropathology/Toxicity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:28294-28304. [PMID: 39661093 DOI: 10.1021/acs.jafc.4c07659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
Extracts from plants/herbals are great resources of drugs and nutrients. Baicalein, a component present in Scutellaria baicalensis, was previously found to alleviate the abnormal depolarization brought about by Aβ oligomers. We extended this promising outcome by screening baicalein derivatives, and a natural compound named homoplantaginin was pinpointed. In this study, we aimed to investigate the effects of homoplantaginin on animal behavior and explore its neuronal functioning/mechanism. In behavior tests, impairments of novel object recognition and of spatial learning/memory were reversed by homoplantaginin in a J20 Alzheimer's disease (AD) mouse model. Utilizing primary glutamatergic neurons, homoplantaginin was found to prevent the Aβ oligomer-induced increase in ERK phosphorylation. Furthermore, homoplantaginin inhibits both AMPA-insult and NMDA-insult depolarization; this was assessed using DiBAC4(3), a membrane potential sensitive dye. Finally, homoplantaginin blocks both Aβ oligomer-induced and NMDA-induced calcium influx, which was assessed by intracellular calcium measurement using Fura2/AM. Interestingly, homoplantaginin immediately blunts the steady state calcium influx caused by NMDA. Taken together, homoplantaginin is capable of inhibiting Aβ oligomer-induced pathophysiology, in particular at the receptor level. This pure compound has great potential to be developed as a clinical therapeutic drug.
Collapse
Affiliation(s)
- Ting-Yu Chen
- Department of Life Science, National Taiwan Normal University, Taipei 116325, Taiwan
| | - Yi-Ru Chen
- Department of Chemistry, National Taiwan Normal University, Taipei 116325, Taiwan
| | - Ming-Lung Hsu
- Department of Life Science, National Taiwan Normal University, Taipei 116325, Taiwan
| | - Yueh-Ting Liao
- Department of Life Science, National Taiwan Normal University, Taipei 116325, Taiwan
| | - Chia-Hsuan Wu
- Department of Life Science, National Taiwan Normal University, Taipei 116325, Taiwan
| | - Chien-An Yao
- Department of Family Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Wen-Chin Yang
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Wenwei Lin
- Department of Chemistry, National Taiwan Normal University, Taipei 116325, Taiwan
| | - Yenshou Lin
- Department of Life Science, National Taiwan Normal University, Taipei 116325, Taiwan
| |
Collapse
|
75
|
Choi A, Kim B, Labriola E, Wiest A, Wang Y, Smith J, Shin H, Jin X, An I, Hong J, Antila H, Thomas S, Bhattarai JP, Beier K, Ma M, Weber F, Chung S. Circuit mechanism underlying fragmented sleep and memory deficits in 16p11.2 deletion mouse model of autism. iScience 2024; 27:111285. [PMID: 39628570 PMCID: PMC11612818 DOI: 10.1016/j.isci.2024.111285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/26/2024] [Accepted: 10/25/2024] [Indexed: 12/06/2024] Open
Abstract
Sleep disturbances are prevalent in children with autism spectrum disorder (ASD). Strikingly, sleep problems are positively correlated with the severity of ASD symptoms, such as memory impairment. However, the neural mechanisms underlying sleep disturbances and cognitive deficits in ASD are largely unexplored. Here, we show that non-rapid eye movement sleep (NREMs) is fragmented in the 16p11.2 deletion mouse model of ASD. The degree of sleep fragmentation is reflected in an increased number of calcium transients in the activity of locus coeruleus noradrenergic (LC-NE) neurons during NREMs. In contrast, optogenetic inhibition of LC-NE neurons and pharmacological blockade of noradrenergic transmission using clonidine consolidate sleep. Furthermore, inhibiting LC-NE neurons restores memory. Finally, rabies-mediated screening of presynaptic neurons reveals altered connectivity of LC-NE neurons with sleep- and memory-regulatory regions in 16p11.2 deletion mice. Our findings identify a crucial role of the LC-NE system in regulating sleep stability and memory in ASD.
Collapse
Affiliation(s)
- Ashley Choi
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bowon Kim
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eleanor Labriola
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alyssa Wiest
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yingqi Wang
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jennifer Smith
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hyunsoo Shin
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xi Jin
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Isabella An
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jiso Hong
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hanna Antila
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Steven Thomas
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Janardhan P. Bhattarai
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kevin Beier
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA 92617, USA
| | - Minghong Ma
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Franz Weber
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shinjae Chung
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
76
|
Parvathy S, Basu B, Surya S, Jose R, Meera V, Riya PA, Jyothi NP, Sanalkumar R, Praz V, Riggi N, Nair BS, Gulia KK, Kumar M, Binukumar BK, James J. TLX3 regulates CGN progenitor proliferation during cerebellum development and its dysfunction can lead to autism. iScience 2024; 27:111260. [PMID: 39628587 PMCID: PMC11612787 DOI: 10.1016/j.isci.2024.111260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/29/2024] [Accepted: 10/23/2024] [Indexed: 12/06/2024] Open
Abstract
Tlx3, a master regulator of the fate specification of excitatory neurons, is primarily known to function in post-mitotic cells. Although we have previously identified TLX3 expression in the proliferating granule neuron progenitors (GNPs) of cerebellum, its primary role is unknown. Here, we demonstrate that the dysfunction of Tlx3 from the GNPs significantly reduced its proliferation through regulating anti-proliferative genes. Consequently, the altered generation of GNPs resulted in cerebellar hypoplasia, patterning defects, granule neuron-Purkinje ratio imbalance, and aberrant synaptic connections in the cerebellum. This altered cerebellar homeostasis manifested into a typical autism-like behavior in mice with motor, and social function disabilities. We also show the presence of TLX3 variants with uncharacterized mutations in human cases of autism spectrum disorder (ASD). Altogether, our study establishes Tlx3 as a critical gene involved in developing GNPs and that its deletion from the early developmental stage culminates in autism.
Collapse
Affiliation(s)
- Surendran Parvathy
- Neuro Stem Cell Biology Laboratory, Neurobiology Division, Rajiv Gandhi Centre for Biotechnology (BRIC-RGCB), Thiruvananthapuram, Kerala 695 014, India
- Research Centre, The University of Kerala, Thiruvananthapuram, Kerala 695 014, India
| | - Budhaditya Basu
- Neuro Stem Cell Biology Laboratory, Neurobiology Division, Rajiv Gandhi Centre for Biotechnology (BRIC-RGCB), Thiruvananthapuram, Kerala 695 014, India
- Regional Centre for Biotechnology (BRIC-RCB), Faridabad, Haryana 121001, India
| | - Suresh Surya
- Neuro Stem Cell Biology Laboratory, Neurobiology Division, Rajiv Gandhi Centre for Biotechnology (BRIC-RGCB), Thiruvananthapuram, Kerala 695 014, India
- Research Centre, The University of Kerala, Thiruvananthapuram, Kerala 695 014, India
| | - Rahul Jose
- Neuro Stem Cell Biology Laboratory, Neurobiology Division, Rajiv Gandhi Centre for Biotechnology (BRIC-RGCB), Thiruvananthapuram, Kerala 695 014, India
- Regional Centre for Biotechnology (BRIC-RCB), Faridabad, Haryana 121001, India
| | - Vadakkath Meera
- Neuro Stem Cell Biology Laboratory, Neurobiology Division, Rajiv Gandhi Centre for Biotechnology (BRIC-RGCB), Thiruvananthapuram, Kerala 695 014, India
- Research Centre, The University of Kerala, Thiruvananthapuram, Kerala 695 014, India
| | - Paul Ann Riya
- Neuro Stem Cell Biology Laboratory, Neurobiology Division, Rajiv Gandhi Centre for Biotechnology (BRIC-RGCB), Thiruvananthapuram, Kerala 695 014, India
- Research Centre, The University of Kerala, Thiruvananthapuram, Kerala 695 014, India
| | - Nair Pradeep Jyothi
- Neuro Stem Cell Biology Laboratory, Neurobiology Division, Rajiv Gandhi Centre for Biotechnology (BRIC-RGCB), Thiruvananthapuram, Kerala 695 014, India
- Research Centre, The University of Kerala, Thiruvananthapuram, Kerala 695 014, India
| | | | - Viviane Praz
- CHUV-Lausanne University Hospital, Rue du Bugnon 46, 1005 Lausanne, Switzerland
| | - Nicolò Riggi
- CHUV-Lausanne University Hospital, Rue du Bugnon 46, 1005 Lausanne, Switzerland
| | - Biju Surendran Nair
- Neuro Stem Cell Biology Laboratory, Neurobiology Division, Rajiv Gandhi Centre for Biotechnology (BRIC-RGCB), Thiruvananthapuram, Kerala 695 014, India
| | - Kamalesh K. Gulia
- Division of Sleep Research, Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (SCTIMST), Trivandrum, Kerala 695012, India
| | - Mukesh Kumar
- Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi 110025, India
| | | | - Jackson James
- Neuro Stem Cell Biology Laboratory, Neurobiology Division, Rajiv Gandhi Centre for Biotechnology (BRIC-RGCB), Thiruvananthapuram, Kerala 695 014, India
- Research Centre, The University of Kerala, Thiruvananthapuram, Kerala 695 014, India
- Regional Centre for Biotechnology (BRIC-RCB), Faridabad, Haryana 121001, India
| |
Collapse
|
77
|
Shea JM, Villeda SA. MICROGLIA AGING IN THE HIPPOCAMPUS ADVANCES THROUGH INTERMEDIATE STATES THAT DRIVE ACTIVATION AND COGNITIVE DECLINE. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.09.588665. [PMID: 38645176 PMCID: PMC11030314 DOI: 10.1101/2024.04.09.588665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
During aging, microglia - the resident macrophages of the brain - exhibit altered phenotypes and contribute to age-related neuroinflammation. While numerous hallmarks of age-related microglia have been elucidated, the progression from homeostasis to dysfunction during the aging process remains unresolved. To bridge this gap in knowledge, we undertook complementary cellular and molecular analyses of microglia in the mouse hippocampus across the adult lifespan and in the experimental aging model of heterochronic parabiosis. Single-cell RNA-Seq and pseudotime analysis revealed age-related transcriptional heterogeneity in hippocampal microglia and identified intermediate states of microglial aging that also emerge following heterochronic parabiosis. We tested the functionality of intermediate stress response states via TGFβ1 and translational states using pharmacological approaches in vitro to reveal their modulation of the progression to an activated state. Furthermore, we utilized single-cell RNA-Seq in conjunction with in vivo adult microglia-specific Tgfb1 conditional genetic knockout mouse models, to demonstrate that microglia advancement through intermediate aging states drives transcriptional inflammatory activation and hippocampal-dependent cognitive decline.
Collapse
Affiliation(s)
- Jeremy M. Shea
- Department of Anatomy, University of California San Francisco, San Francisco, California 94143, USA
| | - Saul A. Villeda
- Department of Anatomy, University of California San Francisco, San Francisco, California 94143, USA
- Department of Physical Therapy and Rehabilitation Science, University of California San Francisco, San Francisco, California 94143, USA
- Bakar Aging Research Institute, University of California San Francisco, San Francisco, California, 94143, USA
| |
Collapse
|
78
|
Zhukov IS, Alnefeesi Y, Krotova NA, Nemets VV, Demin KA, Karpenko MN, Budygin EA, Kanov EV, Kalueff AV, Shabanov PD, Bader M, Alenina N, Gainetdinov RR. Trace amine-associated receptor 1 agonist reduces aggression in brain serotonin-deficient tryptophan hydroxylase 2 knockout rats. Front Psychiatry 2024; 15:1484925. [PMID: 39748904 PMCID: PMC11693706 DOI: 10.3389/fpsyt.2024.1484925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/25/2024] [Indexed: 01/04/2025] Open
Abstract
Introduction Aggression and self-harm disproportionately occur in youths preoccupied with social status tracking. These pathological conditions are linked to a serotonin (5-HT) deficit in the brain. Ablation of 5-HT biosynthesis by tryptophan hydroxylase 2 knockout (TPH2-KO) increases aggression in rodents. Remarkably, deletion of the trace amine-associated receptor 1 (TAAR1) results in the same consequences. Unlike the nuanced dynamics of social status cues in young people, the social ranks of rats mainly advance when they dominate larger opponents in combat. Methods This study explored whether the potent TAAR1 agonist RO5263397 reduces aggression caused by 5-HT depletion, and whether social rank advancement motivates this aggression. The resident-intruder paradigm was applied with larger and smaller intruders to evaluate whether social rank advancement motivates aggressive behaviors in TPH2-KO rats. Results When a smaller intruder was introduced, 5-HT-deficient rats did not differ from wild type littermates. However, when the intruders were larger, the mutants extended their aggressive efforts, refusing to submit. Importantly, RO5263397 selectively abolished this abnormal form of aggression in TPH2-KO rats. Discussion Results supported social rank advancement as the main incentive. These data also suggest that TAAR1 is a promising target for the development of new treatments for aggression; independent data also support this conclusion.
Collapse
Affiliation(s)
- Ilya S. Zhukov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Yazen Alnefeesi
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | | | - Vsevolod V. Nemets
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Konstantin A. Demin
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | | | - Evgeny A. Budygin
- Department of Neurobiology, Sirius University of Science and Technology, Sirius, Russia
| | - Evgeny V. Kanov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- St. Petersburg University Hospital, St. Petersburg State University, St. Petersburg, Russia
| | - Allan V. Kalueff
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Department of Neurobiology, Sirius University of Science and Technology, Sirius, Russia
- Department of Biosciences and Bioinformatics, School of Science, Xi’an Jiaotong-Liverpool University, Suzhou, China
- Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi’an Jiaotong-Liverpool University, Suzhou, China
| | | | - Michael Bader
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Natalia Alenina
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- St. Petersburg University Hospital, St. Petersburg State University, St. Petersburg, Russia
| |
Collapse
|
79
|
de Diego-Otero Y, El Bekay R, García-Guirado F, Sánchez-Salido L, Giráldez-Pérez RM. Apocynin, a Selective NADPH Oxidase (Nox2) Inhibitor, Ameliorates Behavioural and Learning Deficits in the Fragile X Syndrome Mouse Model. Biomedicines 2024; 12:2887. [PMID: 39767793 PMCID: PMC11673502 DOI: 10.3390/biomedicines12122887] [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: 11/19/2024] [Revised: 12/13/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Fragile X Syndrome (FXS) is associated with intellectual disability, hyperactivity, social anxiety and signs of autism. Hyperactivation of NADPH oxidase has been previously described in the brain of the male Fmr1-KO mouse. This work aims to demonstrate the efficacy of Apocynin, a specific NADPH oxidase inhibitor, in treating Fragile X mouse hallmarks. Methods: Free radicals, lipid and protein oxidation markers and behavioural and learning paradigms were measured after chronic treatment with orally administered vehicle, 10 mg/kg/day or 30 mg/kg/day of Apocynin. Results: The results revealed a reduction in testis weight, an increase in peritoneal fat, and no variation in body weight after chronic treatment. Furthermore, a reduction in hyperactivity was detected in Apocynin-treated male Fmr1-KO mice. Additionally, the higher dose of 30 mg/kg/day also improves behaviour and learning in the male Fmr1-KO mice, normalising free radical production and oxidative parameters. Moreover, a reduction in phospho-EKR1 and P47-Phox protein signals was observed in specific brain areas. Conclusions: Thus, chronic treatment with Apocynin could lead to a new therapeutic option for the Fragile X Syndrome.
Collapse
Affiliation(s)
- Yolanda de Diego-Otero
- Cellular Biology, Physiology and Immunology Department, University of Córdoba, 14014 Córdoba, Spain;
| | - Rajaa El Bekay
- Research Laboratory, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIO-NAND, Hospital Civil, 29009 Malaga, Spain
- Endocrinology and Nutrition Clinic Unit, Regional University Hospital of Málaga, 29009 Málaga, Spain
- CIBER of Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Francisco García-Guirado
- Research Laboratory, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIO-NAND, Hospital Civil, 29009 Malaga, Spain
| | - Lourdes Sánchez-Salido
- Research Laboratory, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIO-NAND, Hospital Civil, 29009 Malaga, Spain
| | - Rosa María Giráldez-Pérez
- Cellular Biology, Physiology and Immunology Department, University of Córdoba, 14014 Córdoba, Spain;
| |
Collapse
|
80
|
Rochais C, Lecoutey C, Lalut J, Davis A, Duval E, Gaven F, Largillière S, Née G, Corvaisier S, Sopkova de Oliveira Santos J, Since M, Freret T, Legrand R, Callizot N, Claeysen S, Boulouard M, Dallemagne P. Synthesis, in vitro and in vivo biological evaluation of novel dual compounds targeting both acetylcholinesterase and serotonergic 5-HT 4 receptors with potential interest in the treatment of Alzheimer's disease. Eur J Med Chem 2024; 280:116975. [PMID: 39454222 DOI: 10.1016/j.ejmech.2024.116975] [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: 09/20/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024]
Abstract
In this work, we exemplified the "copride" family of drug candidates able to both inhibit acetylcholinesterase and to activate 5-HT4 receptors, with anti-amnesiant and promnesiant activities in mice. Twenty-one analogs of donecopride, the first-in class representative of the series, were synthesized exploring the influence on the biological activities of the substituents (methoxy, amine and chlorine) carried by its phenyl ring. This work was the support of an intensive structure-activity relationship study and allowed to obtain some interesting derivatives of donecopride. In this respect, the replacement of the methoxy group of the latter with a deuterated one led to deudonecopride. On the other hand, the replacement of the chlorine atom of donecopride by various halogen atoms was of particular interest, among which fluorine led to a potent analog, we called flucopride. The latter exhibited promising in vitro activities associated to excellent drugability parameters. Flucopride was consequently involved in in vivo studies such as a scopolamine-induced deficit model of working memory and in a novel object recognition test. Through these evaluations, flucopride demonstrated both its antiamnesiant and promnesiant capacities, which could make it a potential preclinical drug candidate for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Christophe Rochais
- Université de Caen Normandie, Normandie Univ., Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), 14000 Caen, France.
| | - Cédric Lecoutey
- Université de Caen Normandie, Normandie Univ., Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), 14000 Caen, France
| | - Julien Lalut
- Université de Caen Normandie, Normandie Univ., Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), 14000 Caen, France
| | - Audrey Davis
- Université de Caen Normandie, Normandie Univ., Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), 14000 Caen, France
| | - Emilie Duval
- Université de Caen Normandie, Normandie Univ., Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), 14000 Caen, France
| | - Florence Gaven
- IGF, Univ. Montpellier, CNRS, INSERM, F-34094, Montpellier, France
| | - Stacy Largillière
- Université de Caen Normandie, Normandie Univ., Mobilités: Vieillissement, Pathologie, Santé (COMETE), INSERM UMR-S 1075, 14000, Caen, France
| | - Gérald Née
- Université de Caen Normandie, Normandie Univ., Mobilités: Vieillissement, Pathologie, Santé (COMETE), INSERM UMR-S 1075, 14000, Caen, France
| | - Sophie Corvaisier
- Université de Caen Normandie, Normandie Univ., Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), 14000 Caen, France
| | - Jana Sopkova de Oliveira Santos
- Université de Caen Normandie, Normandie Univ., Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), 14000 Caen, France
| | - Marc Since
- Université de Caen Normandie, Normandie Univ., Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), 14000 Caen, France; PRISMM Platform, PLATON Service Unit, Caen, Université de Caen Normandie, France
| | - Thomas Freret
- Université de Caen Normandie, Normandie Univ., Mobilités: Vieillissement, Pathologie, Santé (COMETE), INSERM UMR-S 1075, 14000, Caen, France
| | - Romain Legrand
- RONOMA Pharma, 31 Rue Léon Delille, F-76800, Saint Etienne du Rouvray, France
| | - Noëlle Callizot
- Neuro-Sys, 410 Chemin départemental 60, F-13120, Gardanne, France
| | - Sylvie Claeysen
- IGF, Univ. Montpellier, CNRS, INSERM, F-34094, Montpellier, France
| | - Michel Boulouard
- Université de Caen Normandie, Normandie Univ., Mobilités: Vieillissement, Pathologie, Santé (COMETE), INSERM UMR-S 1075, 14000, Caen, France
| | - Patrick Dallemagne
- Université de Caen Normandie, Normandie Univ., Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), 14000 Caen, France.
| |
Collapse
|
81
|
Cheng R, Bai N, Liu S, Zhao X, Jiang B, Guo W, Cao S, Liu J, Li N, Li X, Wu X, Yi F, Wang Z, Guo Q, Wei J, Bai M, Jiang X, Song X, Wang Z, Miao Q, Wang D, Di Y, Liu H, Cao L. The deacetylase SIRT6 reduces amyloid pathology and supports cognition in mice by reducing the stability of APP in neurons. Sci Signal 2024; 17:eado1035. [PMID: 39656860 DOI: 10.1126/scisignal.ado1035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 07/10/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024]
Abstract
Alzheimer's disease (AD) is an aging-related neurodegenerative disorder that results in progressively impaired memory and is often associated with amyloid plaques. Previous studies implicate the deacetylases SIRT1 and SIRT2 in regulating the processing of amyloid precursor protein (APP). Here, we investigated whether APP is regulated by the related deacetylase SIRT6, which shows aging-associated decreases in activity. We found that the abundance of SIRT6 was reduced in the cortex and hippocampus of aged and AD model mice and negatively correlated with that of APP. In mouse hippocampal neurons and transfected human cells, SIRT6 interacted with and deacetylated APP at three consecutive Lys residues (Lys649, Lys650, and Lys651). This deacetylation, in turn, increased the ubiquitylation of APP, leading to its proteasomal degradation. SIRT6 abundance in neurons was reduced by oxidative stress and DNA damage, both of which are implicated in neurodegenerative pathology. Systemic pharmacological activation of SIRT6 ameliorated both amyloid pathology and cognitive deficits in APP/PS1 mice, a mouse model of AD. The findings demonstrate that the activity of SIRT6 destabilizes APP and suggest that activating SIRT6 has therapeutic potential to reduce amyloid-associated pathology in patients with AD.
Collapse
Affiliation(s)
- Rong Cheng
- College of Basic Medical Science, Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
- Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, Shenyang, Liaoning 110122, China
| | - Ning Bai
- College of Basic Medical Science, Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
- Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, Shenyang, Liaoning 110122, China
| | - Shuhui Liu
- College of Basic Medical Science, Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
- Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, Shenyang, Liaoning 110122, China
| | - Xiong Zhao
- College of Basic Medical Science, Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
- Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, Shenyang, Liaoning 110122, China
| | - Bo Jiang
- College of Basic Medical Science, Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
- Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, Shenyang, Liaoning 110122, China
| | - Wendong Guo
- College of Basic Medical Science, Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
- Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, Shenyang, Liaoning 110122, China
| | - Sunrun Cao
- College of Basic Medical Science, Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
- Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, Shenyang, Liaoning 110122, China
| | - Jingwei Liu
- College of Basic Medical Science, Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
- Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, Shenyang, Liaoning 110122, China
| | - Na Li
- Department of Gerontology and Geriatrics, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110004, China
| | - Xiaoman Li
- College of Basic Medical Science, Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
- Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, Shenyang, Liaoning 110122, China
| | - Xuan Wu
- College of Basic Medical Science, Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
- Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, Shenyang, Liaoning 110122, China
| | - Fei Yi
- College of Basic Medical Science, Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
- Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, Shenyang, Liaoning 110122, China
| | - Zhuo Wang
- College of Basic Medical Science, Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
- Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, Shenyang, Liaoning 110122, China
| | - Qiqiang Guo
- College of Basic Medical Science, Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
- Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, Shenyang, Liaoning 110122, China
| | - Jiayi Wei
- Department of Developmental Cell Biology, School of Life Sciences; Key Laboratory of Cell Biology, Ministry of Public Health; Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Ming Bai
- College of Basic Medical Science, Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
- Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, Shenyang, Liaoning 110122, China
| | - Xiaoyou Jiang
- College of Basic Medical Science, Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
- Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, Shenyang, Liaoning 110122, China
| | - Xiaoyu Song
- College of Basic Medical Science, Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
- Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, Shenyang, Liaoning 110122, China
| | - Zhuo Wang
- College of Basic Medical Science, Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
- Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, Shenyang, Liaoning 110122, China
| | - Qi Miao
- Innovation Center of Aging-Related Disease Diagnosis and Treatment and Prevention, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
| | - Difei Wang
- Department of Gerontology and Geriatrics, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110004, China
| | - Yu Di
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Hua Liu
- Innovation Center of Aging-Related Disease Diagnosis and Treatment and Prevention, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
| | - Liu Cao
- College of Basic Medical Science, Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
- Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, Shenyang, Liaoning 110122, China
- Innovation Center of Aging-Related Disease Diagnosis and Treatment and Prevention, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
| |
Collapse
|
82
|
Jimenez V, Sacristan V, Jambrina C, Jaen ML, Casana E, Muñoz S, Marcó S, Molas M, Garcia M, Grass I, León X, Elias I, Ribera A, Elias G, Sanchez V, Vilà L, Casellas A, Ferre T, Rodó J, Carretero A, Pumarola M, Navarro M, Andaluz A, Moll X, Añor S, Franckhauser S, Vergara M, Caixàs A, Bosch F. Reversion of metabolic dysfunction-associated steatohepatitis by skeletal muscle-directed FGF21 gene therapy. Mol Ther 2024; 32:4285-4302. [PMID: 39489916 PMCID: PMC11638876 DOI: 10.1016/j.ymthe.2024.10.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/25/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024] Open
Abstract
The highly prevalent metabolic dysfunction-associated steatohepatitis (MASH) is associated with liver steatosis, inflammation, and hepatocyte injury, which can lead to fibrosis and may progress to hepatocellular carcinoma and death. New treatment modalities such as gene therapy may be transformative for MASH patients. Here, we describe that one-time intramuscular administration of adeno-associated viral vectors of serotype 1 (AAV1) encoding native fibroblast growth factor 21 (FGF21), a key metabolic regulator, resulted in sustained increased circulating levels of the factor, which mediated long-term (>1 year) MASH and hepatic fibrosis reversion and halted development of liver tumors in obese male and female mouse models. AAV1-FGF21 treatment also counteracted obesity, adiposity, and insulin resistance, which are significant drivers of MASH. Scale-up to large animals successfully resulted in safe skeletal muscle biodistribution and biological activity in key metabolic tissues. Moreover, as a step toward the clinic, circulating FGF21 levels were characterized in obese, insulin-resistant and MASH patients. Overall, these results underscore the potential of the muscle-directed AAV1-FGF21 gene therapy to treat MASH and support its clinical translation.
Collapse
Affiliation(s)
- Veronica Jimenez
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Victor Sacristan
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Claudia Jambrina
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Maria Luisa Jaen
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Estefania Casana
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Sergio Muñoz
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Sara Marcó
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Maria Molas
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Miquel Garcia
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Ignasi Grass
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Xavier León
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Ivet Elias
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Albert Ribera
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Gemma Elias
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Victor Sanchez
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Laia Vilà
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Alba Casellas
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Tura Ferre
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Jordi Rodó
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Ana Carretero
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Animal Health and Anatomy, School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Marti Pumarola
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Animal Medicine and Surgery, School of Veterinary Medicine, Universitat Autònoma Barcelona, 08193 Bellaterra, Spain
| | - Marc Navarro
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Animal Health and Anatomy, School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Anna Andaluz
- Department of Animal Medicine and Surgery, School of Veterinary Medicine, Universitat Autònoma Barcelona, 08193 Bellaterra, Spain
| | - Xavier Moll
- Department of Animal Medicine and Surgery, School of Veterinary Medicine, Universitat Autònoma Barcelona, 08193 Bellaterra, Spain
| | - Sonia Añor
- Department of Animal Medicine and Surgery, School of Veterinary Medicine, Universitat Autònoma Barcelona, 08193 Bellaterra, Spain
| | - Sylvie Franckhauser
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Mercedes Vergara
- Institut d'Investigació i Innovació Parc Taulí (I3PT-CERCA), 08202 Sabadell, Spain; Department of Medicine, Universitat Autònoma de Barcelona, Teaching Unit Parc Taulí, 08202 Sabadell, Spain; Department of Hepatology, Digestive Service, Hospital Universitari Parc Taulí, 08202 Sabadell, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28029 Madrid, Spain
| | - Assumpta Caixàs
- Institut d'Investigació i Innovació Parc Taulí (I3PT-CERCA), 08202 Sabadell, Spain; Department of Medicine, Universitat Autònoma de Barcelona, Teaching Unit Parc Taulí, 08202 Sabadell, Spain; Department of Endocrinology and Nutrition, Hospital Universitari Parc Taulí, 08202 Sabadell, Spain
| | - Fatima Bosch
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain.
| |
Collapse
|
83
|
Laughlin M, McIndoe R, Adams SH, Araiza R, Ayala JE, Kennedy L, Lanoue L, Lantier L, Macy J, Malabanan E, McGuinness OP, Perry R, Port D, Qi N, Elias CF, Shulman GI, Wasserman DH, Lloyd KCK. The mouse metabolic phenotyping center (MMPC) live consortium: an NIH resource for in vivo characterization of mouse models of diabetes and obesity. Mamm Genome 2024; 35:485-496. [PMID: 39191872 PMCID: PMC11522164 DOI: 10.1007/s00335-024-10067-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: 07/02/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024]
Abstract
The Mouse Metabolic Phenotyping Center (MMPC)Live Program was established in 2023 by the National Institute for Diabetes, Digestive and Kidney Diseases (NIDDK) at the National Institutes of Health (NIH) to advance biomedical research by providing the scientific community with standardized, high quality phenotyping services for mouse models of diabetes and obesity. Emerging as the next iteration of the MMPC Program which served the biomedical research community for 20 years (2001-2021), MMPCLive is designed as an outwardly-facing consortium of service cores that collaborate to provide reduced-cost consultation and metabolic, physiologic, and behavioral phenotyping tests on live mice for U.S. biomedical researchers. Four MMPCLive Centers located at universities around the country perform complex and often unique procedures in vivo on a fee for service basis, typically on mice shipped from the client or directly from a repository or vendor. Current areas of expertise include energy balance and body composition, insulin action and secretion, whole body carbohydrate and lipid metabolism, cardiovascular and renal function, food intake and behavior, microbiome and xenometabolism, and metabolic pathway kinetics. Additionally, an opportunity arose to reduce barriers to access and expand the diversity of the biomedical research workforce by establishing the VIBRANT Program. Directed at researchers historically underrepresented in the biomedical sciences, VIBRANT-eligible investigators have access to testing services, travel and career development awards, expert advice and experimental design consultation, and short internships to learn test technologies. Data derived from experiments run by the Centers belongs to the researchers submitting mice for testing which can be made publicly available and accessible from the MMPCLive database following publication. In addition to services, MMPCLive staff provide expertise and advice to researchers, develop and refine test protocols, engage in outreach activities, publish scientific and technical papers, and conduct educational workshops and training sessions to aid researchers in unraveling the heterogeneity of diabetes and obesity.
Collapse
Affiliation(s)
- Maren Laughlin
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, USA
| | - Richard McIndoe
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, USA
| | - Sean H Adams
- Department of Surgery, School of Medicine, University of California Davis, Davis, USA
- Center for Alimentary and Metabolic Science, School of Medicine, University of California Davis, Davis, USA
| | - Renee Araiza
- Mouse Biology Program, University of California Davis, Davis, USA
| | | | - Lucy Kennedy
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, USA
| | - Louise Lanoue
- Mouse Biology Program, University of California Davis, Davis, USA
| | | | - James Macy
- Department of Comparative Medicine, Yale School of Medicine, New Haven, USA
| | | | | | - Rachel Perry
- Department of Internal Medicine, Yale School of Medicine, New Haven, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, USA
| | - Daniel Port
- Mouse Biology Program, University of California Davis, Davis, USA
| | - Nathan Qi
- Department of Molecular & Integrated Physiology, University of Michigan, Ann Arbor, USA
- Caswell Diabetes Institute, University of Michigan Medical School, Ann Arbor, USA
| | - Carol F Elias
- Department of Molecular & Integrated Physiology, University of Michigan, Ann Arbor, USA
- Caswell Diabetes Institute, University of Michigan Medical School, Ann Arbor, USA
| | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, USA
| | | | - K C Kent Lloyd
- Department of Surgery, School of Medicine, University of California Davis, Davis, USA.
- Mouse Biology Program, University of California Davis, Davis, USA.
| |
Collapse
|
84
|
Franco B, Martineli Rodrigues GA, Rocha VD, Damiani L, Manconi M, Torsoni AS, Esteves AM. Exercise protects impairments in memory recognition in the iron-deficient male rat model of Restless Legs Syndrome. Physiol Behav 2024; 287:114688. [PMID: 39233255 DOI: 10.1016/j.physbeh.2024.114688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024]
Abstract
Restless Legs Syndrome (RLS) is a neurological disorder characterized by an irresistible urge to move the legs and is associated with decreased quality of life and sleep, and may result in cognitive changes. Physical exercise generates cognitive improvements and improves RLS symptoms. Our objective is to analyze recognition memory in the iron-deficient rodent model of RLS, and the effect of exercise. The animals (male Wistar rats) were distributed at 21 days of age into a control group (CTRL) (standard diet) or an ID group (iron-deficient diet). After performance classification (at 77 days of age), the animals were redistributed into CTRL (no exercise), CTRL EX (exercise), ID (no exercise) and ID EX (exercise), totaling 9 animals per group. The exercise groups performed treadmill exercise for four weeks. In the 14th week of the diet, the sleep recording of CTRL and ID animals was carried out to validate the RLS model. The Novel Object Recognition Memory test (NOR) was performed before the start of exercise (8th week of diet) and after the end (14th week) in all groups. The ID group demonstrated worsening sleep parameters and increased paw movements compared to the control group. The ID group demonstrated impairment of recognition memory after 14 weeks of diet compared to the CTRL group, and, the CTRL improved recognition memory in the 14th week compared to the 8th week. No differences were found for the exercise groups. Our findings indicate that the RLS animal model exhibited cognitive alterations associated with recognition memory, and long-term aerobic exercise intervention demonstrated a protective influence against these effects.
Collapse
Affiliation(s)
- Beatriz Franco
- Faculdade de Educação Física, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | | | | | - Luiza Damiani
- Faculdade de Ciências Aplicadas, Universidade Estadual de Campinas, Limeira, SP, Brazil
| | - Mauro Manconi
- Sleep and Epilepsy Center, Neurocenter of Southern Switzerland, Civic Hospital of Lugano (EOC), Lugano, Switzerland
| | - Adriana Souza Torsoni
- Faculdade de Ciências Aplicadas, Universidade Estadual de Campinas, Limeira, SP, Brazil
| | | |
Collapse
|
85
|
Li Y, Yu J, Yang N, Long S, Li Y, Zhao L, Yu Y. Alterations in hippocampal somatostatin interneurons, GABAergic metabolism, and ASL perfusion in an aged male mouse model of POCD aggravated by sleep fragmentation. Physiol Rep 2024; 12:e70153. [PMID: 39648073 PMCID: PMC11625499 DOI: 10.14814/phy2.70153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/29/2024] [Accepted: 11/29/2024] [Indexed: 12/10/2024] Open
Abstract
Sleep fragmentation (SF) is increasingly recognized as a contributing factor to postoperative cognitive dysfunction (POCD). Given the critical roles of somatostatin (SST) interneurons, associated gamma-aminobutyric acid (GABA)ergic neurotransmitters, and hippocampal perfusion in sleep-related cognition, this study examined changes in these mechanisms in preoperative SF affecting POCD induced by anesthesia/surgery in aged male mice. The Morris water maze (MWM), novel object recognition (NOR), and Y maze tests were utilized to evaluate POCD. Arterial spin labeling (ASL) was employed to measure hippocampal regional cerebral blood flow (rCBF). In vitro assays quantified the levels of GABAergic metabolites-such as SST, neuropeptide Y (NPY), glutamic acid decarboxylase 1 (GAD1), vesicular GABA transporter (VGAT), and GABA and the distribution of SST interneurons in the hippocampus through enzyme-linked immunosorbent assay and immunofluorescence. Preoperative 24-h SF exacerbated anesthesia/surgery-induced spatial memory impairments observed in the MWM, NOR, and Y maze tests. Preoperative 24-h SF significantly increased the number of SST interneurons in hippocampal CA1, elevated hippocampal levels of SST, NPY, GAD1, and GABA, and reduced the rCBF. Preoperative SF aggravated POCD in aged male mice, with an increased number of SST interneurons in hippocampal CA1, elevated hippocampal GABAergic metabolites, and a further reduction in rCBF.
Collapse
Affiliation(s)
- Yun Li
- Department of AnesthesiologyTianjin Medical University General HospitalTianjinChina
- Tianjin Research Institute of AnesthesiologyTianjinChina
| | - Jiafeng Yu
- Department of AnesthesiologyTianjin Medical University General HospitalTianjinChina
- Tianjin Research Institute of AnesthesiologyTianjinChina
| | - Ningzhi Yang
- Department of AnesthesiologyTianjin Medical University General HospitalTianjinChina
- Tianjin Research Institute of AnesthesiologyTianjinChina
| | - Siwen Long
- Department of AnesthesiologyTianjin Medical University General HospitalTianjinChina
- Tianjin Research Institute of AnesthesiologyTianjinChina
| | - Yize Li
- Department of AnesthesiologyTianjin Medical University General HospitalTianjinChina
- Tianjin Research Institute of AnesthesiologyTianjinChina
| | - Lina Zhao
- Department of Critical Care MedicineTianjin Medical University General HospitalTianjinChina
| | - Yonghao Yu
- Department of AnesthesiologyTianjin Medical University General HospitalTianjinChina
- Tianjin Research Institute of AnesthesiologyTianjinChina
| |
Collapse
|
86
|
Cheng J, Williams JP, Zhou L, Wang PC, Sun LN, Li RH, An JX. Ozone rectal insufflation mitigates chronic rapid eye movement sleep deprivation-induced cognitive impairment through inflammation alleviation and gut microbiota regulation in mice. Med Gas Res 2024; 14:213-224. [PMID: 39073330 DOI: 10.4103/mgr.medgasres-d-23-00036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 10/20/2023] [Indexed: 07/30/2024] Open
Abstract
A range of sleep disorders has the potential to adversely affect cognitive function. This study was undertaken with the objective of investigating the effects of ozone rectal insufflation (O3-RI) on cognitive dysfunction induced by chronic REM sleep deprivation, as well as elucidating possible underlying mechanisms. O3-RI ameliorated cognitive dysfunction in chronic REM sleep deprived mice, improved the neuronal damage in the hippocampus region and decreased neuronal loss. Administration of O3-RI may protect against chronic REM sleep deprivation induced cognitive dysfunction by reversing the abnormal expression of Occludin and leucine-rich repeat and pyrin domain-containing protein 3 inflammasome as well as interleukin-1β in the hippocampus and colon tissues. Moreover, the microbiota diversity and composition of sleep deprivation mice were significantly affected by O3-RI intervention, as evidenced by the reversal of the Firmicutes/Bacteroidetes abundance ratio and the relative abundance of the Bacteroides genus. In particular, the relative abundance of the Bacteroides genus demonstrated a pronounced correlation with cognitive impairment and inflammation. Our findings suggested that O3-RI can improve cognitive dysfunction in sleep deprivation mice, and its mechanisms may be related to regulating gut microbiota and alleviating inflammation and damage in the hippocampus and colon.
Collapse
Affiliation(s)
- Jie Cheng
- Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - John P Williams
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Li Zhou
- Institute for lnnoration Diagnosis & Treatment in Anesthesiology, School of Anesthesiology, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Peng-Cheng Wang
- Institute for lnnoration Diagnosis & Treatment in Anesthesiology, School of Anesthesiology, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Li-Na Sun
- Institute for lnnoration Diagnosis & Treatment in Anesthesiology, School of Anesthesiology, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Rui-Hua Li
- Institute for lnnoration Diagnosis & Treatment in Anesthesiology, School of Anesthesiology, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Jian-Xiong An
- Medical School, University of Chinese Academy of Sciences, Beijing, China
- Institute for lnnoration Diagnosis & Treatment in Anesthesiology, School of Anesthesiology, Shandong Second Medical University, Weifang, Shandong Province, China
- Center of Anesthesiology, Pain and Sleep Medicine, Rapid Anti-depression, The Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong Province, China
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
87
|
Shirokov A, Zlatogosrkaya D, Adushkina V, Vodovozova E, Kardashevskaya K, Sultanov R, Kasyanov S, Blokhina I, Terskov A, Tzoy M, Evsyukova A, Dubrovsky A, Tuzhilkin M, Elezarova I, Dmitrenko A, Manzhaeva M, Krupnova V, Semiachkina-Glushkovskaia A, Ilyukov E, Myagkov D, Tuktarov D, Popov S, Inozemzev T, Navolokin N, Fedosov I, Semyachkina-Glushkovskaya O. Plasmalogens Improve Lymphatic Clearance of Amyloid Beta from Mouse Brain and Cognitive Functions. Int J Mol Sci 2024; 25:12552. [PMID: 39684263 DOI: 10.3390/ijms252312552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/16/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Amyloid beta (Aβ) is a neuronal metabolic product that plays an important role in maintaining brain homeostasis. Normally, intensive brain Aβ formation is accompanied by its effective lymphatic removal. However, the excessive accumulation of brain Aβ is observed with age and during the development of Alzheimer's disease (AD) leading to cognitive impairment and memory deficits. There is emerging evidence that plasmalogens (Pls), as one of the key brain lipids, may be beneficial for AD and cognitive aging. Here, we studied the effects of Pls on cognitive functions and the lymphatic clearance of Aβ from the brain of AD mice and mice of different ages. The results showed that Pls effectively reduce brain Aβ levels and facilitate learning in aged but not old mice. In AD mice, Pls improve the lymphatic clearance of Aβ that is accompanied by an increase in general motor activity and an improvement of the emotional status and learning ability. Thus, these findings suggest that Pls could be a promising candidate for the alternative or concomitant therapy of AD and age-related brain diseases to enhance the lymphatic clearance of Aβ from the brain and cognitive functions.
Collapse
Affiliation(s)
- Alexander Shirokov
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Russian Academy of Sciences, Prospekt Entuziastov 13, 410049 Saratov, Russia
- Department of Biology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia
| | - Daria Zlatogosrkaya
- Department of Biology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia
| | - Viktoria Adushkina
- Department of Biology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia
| | - Elena Vodovozova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ul. Miklukho-Maklaya, 16/10, 117997 Moscow, Russia
| | - Kristina Kardashevskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ul. Miklukho-Maklaya, 16/10, 117997 Moscow, Russia
| | - Ruslan Sultanov
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Palchevskogo Str. 17, 690041 Vladivostok, Russia
| | - Sergey Kasyanov
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Palchevskogo Str. 17, 690041 Vladivostok, Russia
| | - Inna Blokhina
- Department of Biology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia
| | - Andrey Terskov
- Department of Biology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia
| | - Maria Tzoy
- Physics Department, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia
| | - Arina Evsyukova
- Department of Biology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia
| | - Alexander Dubrovsky
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ul. Miklukho-Maklaya, 16/10, 117997 Moscow, Russia
| | - Matvey Tuzhilkin
- Department of Biology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia
| | - Inna Elezarova
- Department of Biology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia
| | - Alexander Dmitrenko
- Department of Biology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia
| | - Maria Manzhaeva
- Department of Biology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia
| | - Valeria Krupnova
- Department of Biology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia
| | | | - Egor Ilyukov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ul. Miklukho-Maklaya, 16/10, 117997 Moscow, Russia
| | - Dmitry Myagkov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ul. Miklukho-Maklaya, 16/10, 117997 Moscow, Russia
| | - Dmitry Tuktarov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ul. Miklukho-Maklaya, 16/10, 117997 Moscow, Russia
| | - Sergey Popov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ul. Miklukho-Maklaya, 16/10, 117997 Moscow, Russia
| | - Tymophey Inozemzev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ul. Miklukho-Maklaya, 16/10, 117997 Moscow, Russia
| | - Nikita Navolokin
- Department of Biology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia
- Department of Pathological Anatomy, Saratov Medical State University, Bolshaya Kazachaya Str. 112, 410012 Saratov, Russia
| | - Ivan Fedosov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ul. Miklukho-Maklaya, 16/10, 117997 Moscow, Russia
| | | |
Collapse
|
88
|
Rakshit D, Goyal R, Yadav V, Gore SK, Sen S, Ranjan OP, Mishra A. Nanoformulated fisetin ameliorates Alzheimer's disease via reducing proinflammatory cytokines and activating the NRF2/HO-1 pathway. Nanomedicine (Lond) 2024; 19:2537-2553. [PMID: 39552578 DOI: 10.1080/17435889.2024.2419814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/18/2024] [Indexed: 11/19/2024] Open
Abstract
Aim: The study aimed to evaluate the neuroprotective effect of a chitosan-coated fisetin nanoformulation in an experimental Alzheimer's disease (AD) model, focusing on improving fisetin's pharmacokinetics and exploring its impact on both brain and colon pathology.Materials & methods: AD was induced in mice by intracerebroventricular administration of Aβ1-42. Mice were treated with either fisetin or a fisetin nanoformulation (5 mg/kg/day, orally) for 21 days. Behavioural assessments were conducted to evaluate memory impairment, motor deficits, and depression-like behaviour. Oxidative stress markers and pro-inflammatory cytokines were measured in the cortex, hippocampus and colon. The changes in cortical and hippocampal AChE levels were also recorded. Histological studies were performed on the cortex, hippocampus (dentate gyrus), and proximal colon.Results: The fisetin nanoformulation significantly improved neurobehavioral outcomes, reducing memory impairment, motor deficits and depression-like symptoms induced by Aβ1-42. It also decreased oxidative and nitrosative stress, along with pro-inflammatory cytokine levels in the cortex, hippocampus and colon. Histological analyses revealed improved brain and colon tissue architecture after treatment with the nanoformulation.Conclusion: The chitosan-coated fisetin nanoformulation enhanced the neuroprotective effects of fisetin in an AD model, likely by improving its pharmacokinetic profile. The findings also suggest a potential link between colon health and Aβ-induced AD pathology, underscoring the therapeutic potential of fisetin nanoformulations in AD management.
Collapse
Affiliation(s)
- Debarati Rakshit
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research (NIPER) - Guwahati, Changsari, Kamrup, Assam, 781101, India
| | - Ritish Goyal
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research (NIPER) - Guwahati, Changsari, Kamrup, Assam, 781101, India
| | - Vikas Yadav
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research (NIPER) - Guwahati, Changsari, Kamrup, Assam, 781101, India
| | - Swati Kailas Gore
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education & Research (NIPER) - Guwahati, Changsari, Kamrup, Assam, 781101, India
| | - Srijita Sen
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education & Research (NIPER) - Guwahati, Changsari, Kamrup, Assam, 781101, India
| | - Om Prakash Ranjan
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education & Research (NIPER) - Guwahati, Changsari, Kamrup, Assam, 781101, India
| | - Awanish Mishra
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research (NIPER) - Guwahati, Changsari, Kamrup, Assam, 781101, India
| |
Collapse
|
89
|
Jiang J, Zhang P, Yuan Y, Xu X, Wu T, Zhang Z, Wang J, Bi Y. Prolactin deficiency drives diabetes-associated cognitive dysfunction by inducing microglia-mediated synaptic loss. J Neuroinflammation 2024; 21:295. [PMID: 39543619 PMCID: PMC11566644 DOI: 10.1186/s12974-024-03289-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Diabetes-associated cognitive dysfunction, characterized by hippocampal synaptic loss as an early pathological feature, seriously threatens patients' quality of life. Synapses are dynamic structures, and hormones play important roles in modulating the formation and elimination of synapses. The pituitary, the master gland of the body, releases several hormones with multiple roles in hippocampal synaptic regulation. In this study, we aimed to explore the relationship between pituitary hormones and cognitive decline in diabetes. METHODS A total of 744 patients with type 2 diabetes (T2DM) (445 men and 299 postmenopausal women) who underwent serum pituitary hormone level assessments, comprehensive cognitive evaluations and MRI scans were enrolled. Dynamic diet interventions were applied in both chow diet-fed mice and high-fat diet (HFD)-fed diabetic mice. The cognitive performance and hippocampal pathology of prolactin (PRL)-knockout mice, neuronal prolactin receptor (PRLR)-specific knockout mice and microglial PRLR-specific knockout mice were assessed. Microglial PRLR-specific knockout mice were fed an HFD to model diabetes. Diabetic mice received an intracerebroventricular infusion of recombinant PRL protein or vehicle. RESULTS This clinical study revealed that decreased PRL levels were associated with cognitive impairment and hippocampal damage in T2DM patients. In diabetic mice, PRL levels diminished before hippocampal synaptic loss and cognitive decline occurred. PRL loss could directly cause cognitive dysfunction and decreased hippocampal synaptic density. Knockout of PRLR in microglia, rather than neurons, induced hippocampal synaptic loss and cognitive impairment. Furthermore, blockade of PRL/PRLR signaling in microglia exacerbated abnormal microglial phagocytosis of synapses, further aggravating hippocampal synaptic loss and cognitive impairment in diabetic mice. Moreover, PRL infusion reduced microglia-mediated synaptic loss, thereby alleviating cognitive impairment in diabetic mice. CONCLUSION PRL is associated with cognitive dysfunction and hippocampal damage in T2DM patients. In diabetes, a decrease in PRL level drives hippocampal synaptic loss and cognitive impairment by increasing microglia-mediated synapse engulfment. Restoration of PRL levels ameliorates cognitive dysfunction and hippocampal synaptic loss in diabetic mice.
Collapse
Affiliation(s)
- Jiaxuan Jiang
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Pengzi Zhang
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Yue Yuan
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Xiang Xu
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Tianyu Wu
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Zhou Zhang
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China.
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China.
| | - Jin Wang
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China.
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China.
| | - Yan Bi
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China.
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China.
| |
Collapse
|
90
|
Helbling JC, Ginieis R, Mortessagne P, Ruiz-Gayo M, Bakoyiannis I, Ducourneau EG, Ciocca D, Bouleté IM, Favereaux A, Ces A, Montalban E, Capuron L, Jeanneteau F, Ferreira G, Challet E, Moisan MP. Time-restricted feeding prevents memory impairments induced by obesogenic diet consumption, via hippocampal thyroid hormone signaling. Mol Metab 2024; 90:102061. [PMID: 39515608 DOI: 10.1016/j.molmet.2024.102061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/21/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVE The early consumption of calorie-rich diet disrupts circadian rhythms and has adverse effects on memory, yet the effects of time-restricted feeding (TRF) and the underlying molecular mechanisms are unknown. Here, we set out to identify the behavioral and molecular circadian rhythms disruptions generated by juvenile obesogenic diet consumption and their restoration by TRF in male mice. METHODS Metabolic rhythms were measured by indirect calorimetry and memory performances by behavioral tasks. Hippocampal translatome (pS6_TRAP), enrichment and co-regulated gene network analyses were conducted to identify the molecular pathways involved in memory impairments and their restoration by TRF. Differential exon usage analyses, mass spectrometry and pharmacological intervention were used to confirm thyroid hormone signaling involvement. RESULTS We show that four weeks of TRF restore the rhythmicity of metabolic parameters and prevents memory impairments in mice fed a high fat-high sucrose (HFS) diet since weaning, independently of body fat levels. Hippocampal translatome and differential exon usage analyses indicate that impaired memory of mice under ad libitum HFS diet is accompanied by reduced thyroid hormone signaling and altered expression of astrocytic genes regulating glutamate neurotransmission. TRF restored the diurnal expression variation of part of these genes and intra-hippocampal infusion of T3, the active form of thyroid hormone, rescues memory performances and astrocytic gene expression of ad libitum HFS diet-fed mice. CONCLUSIONS Thus, thyroid hormones contribute to the TRF positive effects on both metabolism and memory in mice fed an obesogenic diet, highlighting this nutritional approach as a powerful tool in addressing obesity brain comorbidities and paving the way for further mechanistic studies on hippocampal thyroid signaling.
Collapse
Affiliation(s)
- Jean-Christophe Helbling
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Teams NutriPsy & FoodCircus, Bordeaux, France
| | - Rachel Ginieis
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Teams NutriPsy & FoodCircus, Bordeaux, France
| | - Pierre Mortessagne
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Teams NutriPsy & FoodCircus, Bordeaux, France
| | - Mariano Ruiz-Gayo
- Department of Health and Pharmaceutical Sciences, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Ioannis Bakoyiannis
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Teams NutriPsy & FoodCircus, Bordeaux, France
| | - Eva-Gunnel Ducourneau
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Teams NutriPsy & FoodCircus, Bordeaux, France
| | - Dominique Ciocca
- Chronobiotron, Centre National de la Recherche Scientifique (CNRS), University of Strasbourg, France
| | - Illona-Marie Bouleté
- Chronobiotron, Centre National de la Recherche Scientifique (CNRS), University of Strasbourg, France
| | - Alexandre Favereaux
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Aurélia Ces
- Institute of Cellular and Integrative Neurosciences, CNRS, University of Strasbourg, France
| | - Enrica Montalban
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Teams NutriPsy & FoodCircus, Bordeaux, France
| | - Lucile Capuron
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Teams NutriPsy & FoodCircus, Bordeaux, France
| | - Freddy Jeanneteau
- Institut de Génomique Fonctionnelle, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Guillaume Ferreira
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Teams NutriPsy & FoodCircus, Bordeaux, France
| | - Etienne Challet
- Institute of Cellular and Integrative Neurosciences, CNRS, University of Strasbourg, France
| | - Marie-Pierre Moisan
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Teams NutriPsy & FoodCircus, Bordeaux, France.
| |
Collapse
|
91
|
Abstract
Memories are stored as ensembles of engram neurons and their successful recall involves the reactivation of these cellular networks. However, significant gaps remain in connecting these cell ensembles with the process of forgetting. Here, we utilized a mouse model of object memory and investigated the conditions in which a memory could be preserved, retrieved, or forgotten. Direct modulation of engram activity via optogenetic stimulation or inhibition either facilitated or prevented the recall of an object memory. In addition, through behavioral and pharmacological interventions, we successfully prevented or accelerated forgetting of an object memory. Finally, we showed that these results can be explained by a computational model in which engrams that are subjectively less relevant for adaptive behavior are more likely to be forgotten. Together, these findings suggest that forgetting may be an adaptive form of engram plasticity which allows engrams to switch from an accessible state to an inaccessible state.
Collapse
Affiliation(s)
- James D O'Leary
- School of Biochemistry and Immunology, Trinity College DublinDublinIreland
- Trinity College Institute of Neuroscience, Trinity College DublinDublinIreland
| | - Rasmus Bruckner
- Trinity College Institute of Neuroscience, Trinity College DublinDublinIreland
- Department of Education and Psychology, Freie Universität BerlinBerlinGermany
- Max Planck Research Group NeuroCode, Max Planck Institute for Human DevelopmentBerlinGermany
| | - Livia Autore
- School of Biochemistry and Immunology, Trinity College DublinDublinIreland
- Trinity College Institute of Neuroscience, Trinity College DublinDublinIreland
| | - Tomás J Ryan
- School of Biochemistry and Immunology, Trinity College DublinDublinIreland
- Trinity College Institute of Neuroscience, Trinity College DublinDublinIreland
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of MelbourneMelbourneAustralia
- Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR)TorontoCanada
| |
Collapse
|
92
|
Park SY, Kim KY, Gwak DS, Shin SY, Jun DY, Kim YH. L-Cysteine mitigates ROS-induced apoptosis and neurocognitive deficits by protecting against endoplasmic reticulum stress and mitochondrial dysfunction in mouse neuronal cells. Biomed Pharmacother 2024; 180:117538. [PMID: 39393330 DOI: 10.1016/j.biopha.2024.117538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/30/2024] [Accepted: 10/04/2024] [Indexed: 10/13/2024] Open
Abstract
Oxidative stress and mitochondrial dysfunction play critical roles in neurodegenerative diseases. Glutathione (GSH), a key brain antioxidant, helps to neutralize reactive oxygen species (ROS) and maintain redox balance. We investigated the effectiveness of L-cysteine (L-Cys) in preventing apoptosis induced by the ROS generator 2,3-dimethoxy-1,4-naphthoquinone (DMNQ) in mouse hippocampal neuronal HT22 cells, as well as alleviating memory and cognitive impairments caused by the GSH synthesis inhibitor L-buthionine sulfoximine (BSO) in mice. DMNQ-induced apoptotic events in HT22 cells, including elevated cytosolic and mitochondrial ROS levels, DNA fragmentation, endoplasmic reticulum stress, and mitochondrial damage-mediated apoptotic pathways were dose-dependently abrogated by L-Cys (0.5-2 mM). The reduced intracellular GSH level, caused by DMNQ treatment, was restored by L-Cys cotreatment. Although L-Cys did not significantly restore GSH in the presence of BSO, it prevented DMNQ-induced ROS elevation, mitochondrial damage, and apoptosis. Furthermore, compared to N-acetylcysteine and GSH, L-Cys had higher 2,2-diphenyl-1-picrylhydrazyl and 2,2-azino-bis-3-ethylbenzothiazoline-6-sulphonic acid radical-scavenging activity. L-Cys also restored mitochondrial respiration capacity in DMNQ-treated HT22 cells by reversing mitochondrial fission-fusion dynamic balance. BSO administration (500 mg/kg/day) in mice led to neuronal deficits, including memory and cognitive impairments, which were effectively mitigated by oral L-Cys (15 or 30 mg/kg/day). L-Cys also reduced BSO-induced ROS levels in the mice hippocampus and cortex. These findings suggest that even though it does not contribute to intracellular GSH synthesis, exogenous L-Cys protects neuronal cells against oxidative stress-induced mitochondrial damage and apoptosis, by acting as a ROS scavenger, which is beneficial in ameliorating neurocognitive deficits caused by oxidative stress.
Collapse
Affiliation(s)
- Shin Young Park
- Laboratory of Immunobiology, School of Life Science, College of Natural Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea; AT-31 BIO Inc., Business Incubation Center, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Ki Yun Kim
- Laboratory of Immunobiology, School of Life Science, College of Natural Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea; AT-31 BIO Inc., Business Incubation Center, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Dong Seol Gwak
- Laboratory of Immunobiology, School of Life Science, College of Natural Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Soon Young Shin
- Department of Biological Sciences, Sanghuh College of Life Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Do Youn Jun
- AT-31 BIO Inc., Business Incubation Center, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Young Ho Kim
- Laboratory of Immunobiology, School of Life Science, College of Natural Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea; AT-31 BIO Inc., Business Incubation Center, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea.
| |
Collapse
|
93
|
Pushpam M, Talukdar A, Anilkumar S, Maurya SK, Issac TG, Diwakar L. Recurrent endothelin-1 mediated vascular insult leads to cognitive impairment protected by trophic factor pleiotrophin. Exp Neurol 2024; 381:114938. [PMID: 39197707 DOI: 10.1016/j.expneurol.2024.114938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/12/2024] [Accepted: 08/24/2024] [Indexed: 09/01/2024]
Abstract
Vascular dementia (VaD) is a complex neurodegenerative condition, with cerebral small vessel dysfunctions as the central role in its pathogenesis. Given the lack of suitable animal models to study the disease pathogenesis, we developed a mouse model to closely emulate the clinical scenarios of recurrent transient ischemic attacks (TIAs) leading to VaD using vasoconstricting peptide Endothelin-1(ET-1). We observed that administration of ET-1 led to blood-brain barrier (BBB) disruption and detrimental changes in its components, such as endothelial cells and pericytes, along with neuronal loss and synaptic dysfunction, resulting in irreversible memory loss. Further, in our pursuit of understanding potential interventions, we co-administered pleiotrophin (PTN) alongside ET-1 injections. PTN exhibited remarkable efficacy in preserving vital components of the BBB, including endothelial cells and pericytes, thereby restoring BBB integrity, preventing neuronal loss, and enhancing memory function. Our findings give a valuable framework for understanding the detrimental effects of multiple TIAs on brain health and provide a useful animal model to explore VaD's underlying mechanisms further and pave the way for promising therapies.
Collapse
Affiliation(s)
- Mayank Pushpam
- Centre for Brain Research, Indian Institute of Science, Bangalore 560012, India; Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | - Ankita Talukdar
- Centre for Brain Research, Indian Institute of Science, Bangalore 560012, India
| | - Shobha Anilkumar
- Centre for Brain Research, Indian Institute of Science, Bangalore 560012, India
| | | | - Thomas Gregor Issac
- Centre for Brain Research, Indian Institute of Science, Bangalore 560012, India
| | - Latha Diwakar
- Centre for Brain Research, Indian Institute of Science, Bangalore 560012, India.
| |
Collapse
|
94
|
Hu FF, Pan SY, Chu JY, Liu JJ, Duan TT, Luo Y, Zhou W, Wang ZM, Liu W, Zeng Y. Xanthohumol Protects Against Neuronal Excitotoxicity and Mitochondrial Dysfunction in APP/PS1 Mice: An Omics-Based Study. Nutrients 2024; 16:3754. [PMID: 39519590 PMCID: PMC11548031 DOI: 10.3390/nu16213754] [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: 10/10/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Background: Neuronal excitotoxicity and metabolic decline, which begin in the early stages of Alzheimer's disease (AD), pose challenges for effective amelioration. Our previous work suggested that the natural compound xanthohumol, the most abundant prenylated flavonoid in hops, prevents memory deficits in APP/PS1 mice; however, the underlying mechanisms remain unclear. Methods: This study utilized APP/PS1 mice and cutting-edge omics techniques to investigate the effects of xanthohumol on hippocampal proteome, serum metabolome, and microbiome. Results: Our findings revealed that xanthohumol reduces the postsynaptic overexpression of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, N-methyl-D-aspartate, and metabotropic glutamate receptors, but enhances ATP synthesis and mitophagy in the young AD hippocampus. Further mechanistic analyses suggested systemic regulatory effects, particularly on the decreasing glutamate synthesis in the blood and intestines of AD mice following xanthohumol administration. Conclusions: These results underscore the potential of xanthohumol in mitigating AD pathology through multifaceted mechanisms, sparking interest and curiosity in its preventive and therapeutic potential in AD.
Collapse
Affiliation(s)
- Fei-Fei Hu
- Hubei Provincial Clinical Research Center for Alzheimer’s Disease, Wuhan University of Science and Technology, Wuhan 430065, China; (F.-F.H.); (S.-Y.P.); (J.-Y.C.); (T.-T.D.); (Y.L.); (W.Z.); (Z.-M.W.)
- Brain Science and Advanced Technology Institute, Medical School, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Shi-Yao Pan
- Hubei Provincial Clinical Research Center for Alzheimer’s Disease, Wuhan University of Science and Technology, Wuhan 430065, China; (F.-F.H.); (S.-Y.P.); (J.-Y.C.); (T.-T.D.); (Y.L.); (W.Z.); (Z.-M.W.)
- Brain Science and Advanced Technology Institute, Medical School, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Jin-Yu Chu
- Hubei Provincial Clinical Research Center for Alzheimer’s Disease, Wuhan University of Science and Technology, Wuhan 430065, China; (F.-F.H.); (S.-Y.P.); (J.-Y.C.); (T.-T.D.); (Y.L.); (W.Z.); (Z.-M.W.)
- Brain Science and Advanced Technology Institute, Medical School, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Jian-Jun Liu
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China;
| | - Ting-Ting Duan
- Hubei Provincial Clinical Research Center for Alzheimer’s Disease, Wuhan University of Science and Technology, Wuhan 430065, China; (F.-F.H.); (S.-Y.P.); (J.-Y.C.); (T.-T.D.); (Y.L.); (W.Z.); (Z.-M.W.)
- Brain Science and Advanced Technology Institute, Medical School, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yu Luo
- Hubei Provincial Clinical Research Center for Alzheimer’s Disease, Wuhan University of Science and Technology, Wuhan 430065, China; (F.-F.H.); (S.-Y.P.); (J.-Y.C.); (T.-T.D.); (Y.L.); (W.Z.); (Z.-M.W.)
- Brain Science and Advanced Technology Institute, Medical School, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Wen Zhou
- Hubei Provincial Clinical Research Center for Alzheimer’s Disease, Wuhan University of Science and Technology, Wuhan 430065, China; (F.-F.H.); (S.-Y.P.); (J.-Y.C.); (T.-T.D.); (Y.L.); (W.Z.); (Z.-M.W.)
- Brain Science and Advanced Technology Institute, Medical School, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Zhi-Ming Wang
- Hubei Provincial Clinical Research Center for Alzheimer’s Disease, Wuhan University of Science and Technology, Wuhan 430065, China; (F.-F.H.); (S.-Y.P.); (J.-Y.C.); (T.-T.D.); (Y.L.); (W.Z.); (Z.-M.W.)
- Brain Science and Advanced Technology Institute, Medical School, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Wei Liu
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China;
| | - Yan Zeng
- Hubei Provincial Clinical Research Center for Alzheimer’s Disease, Wuhan University of Science and Technology, Wuhan 430065, China; (F.-F.H.); (S.-Y.P.); (J.-Y.C.); (T.-T.D.); (Y.L.); (W.Z.); (Z.-M.W.)
- Brain Science and Advanced Technology Institute, Medical School, Wuhan University of Science and Technology, Wuhan 430065, China
| |
Collapse
|
95
|
Ren H, Tang L, Yuan Z, Liu Y, Zhou X, Xiao X, Wu X, Chen W, Chen Y, Wang H, Xue Q, Xu X. Combined administration of catalpol, puerarin, gastrodin, and borneol modulates the Tlr4/Myd88/NF-κB signaling pathway and alleviates microglia inflammation in Alzheimer's disease. Front Pharmacol 2024; 15:1492237. [PMID: 39545064 PMCID: PMC11560463 DOI: 10.3389/fphar.2024.1492237] [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: 09/10/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024] Open
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disorder affecting millions of people worldwide, with no effective treatment currently available. In recent decades, various traditional Chinese medicines (TCMs) and their active ingredients have shown the potential to attenuate the pathogenesis of AD in cellular and animal models. However, the effects of TCM formulas, which are typically administered in practice, have been less studied. This study aims to investigate the therapeutic effects of several formulas consisting of 4 components herbal components: catalpol, puerarin, gastrodin, and borneol, on streptozotocin (STZ)-induced AD models in cells and rats. The new object recognition (NOR), elevated plus maze (EMP), and Morris water maze (MWM) tests were used to evaluate the cognitive functions of rats. Golgi staining, Haematoxylin and Eosin (HE) staining, and Nissl staining analyses were employed assess the physiology of hippocampal tissues. Gene expression profiles were analyzed used transcriptomics and reverse transcription quantitative polymerase chain reaction analysis, while protein expression levels were determined using immunoblotting, immunohistochemical, and immunofluorescence. The production of cytokines was evaluated with enzyme-linked immunosorbent assay. The results demonstrated that the combined administration of these 4 components (CPGB) had superior mitigating effects on AD cell model, as evidenced by the reduced pro-inflammatory cytokine production and decreased deposition of Aβ protein. Further in vivo and in vitro experiments confirmed that varying doses of CPGB formula effectively ameliorated STZ-induced cognitive deficits, as shown by NOR, MWM, and EMP tests, as well as pathological changes in hippocampal tissues and a 3-dimensional brain neurovascular unit (3D-NVU) model, including decreased deposition of Aβ protein and formation of plaques. Transcriptome sequencing and analysis identified 35 genes with significantly altered expression levels due to STZ and CPGB treatment in hippocampal tissues, which were enriched in the Tlr4/Myd88/NF-κB signaling pathway. Interference with this pathway significantly influenced the therapeutic effects of CPGB in the 3D-NVU model. Collectively, these findings suggest that the combined administration of catalpol, puerarin, gastrodin, and borneol offers superior therapeutic effects on AD by modulating the Tlr4/Myd88/NF-κB signaling pathway. This study strengthens the theoretical foundation for using TCMs to treat AD, proving new insights and references for alleviating and treating AD.
Collapse
Affiliation(s)
- Huijing Ren
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
- Tongren Polytechnic College, Tongren, Guizhou, China
| | - Ling Tang
- Shapingba District People’s Hospital of Chongqing, Chongqing, China
| | - Zhiying Yuan
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Yang Liu
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Xuejiao Zhou
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Xiao Xiao
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Xingyu Wu
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Weihai Chen
- Faculty of Psychology, Southwest University, Chongqing, China
| | - Yi Chen
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Hongjin Wang
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Qiang Xue
- Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Xiaoyu Xu
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
- Southwest University Hospital, Chongqing, China
- Chongqing Key Laboratory of New Drug Screening form Traditional Chinese Medicine, Chongqing, China
- Key Disciplines of Traditional Chinese Medicine of Chongqing City, Rehabilitation Medicine of Southwest University, Chongqing, China
| |
Collapse
|
96
|
Wang X, Chen H, Tang T, Zhan X, Qin S, Hang T, Song M. Chronic Sleep Deprivation Altered the Expression of Memory-Related Genes and Caused Cognitive Memory Dysfunction in Mice. Int J Mol Sci 2024; 25:11634. [PMID: 39519186 PMCID: PMC11546330 DOI: 10.3390/ijms252111634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/18/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Lack of sleep, whether acute or chronic, is quite common and negatively affects an individual's memory and cognitive function. The question of whether chronic sleep deprivation (CSD) causes cognitive impairment to arise and progress is not well studied. To investigate the effects of CSD on memory and cognition, this study began by establishing a CSD mouse model. Behavioral experiments on animals revealed that CSD induced cognitive behavioral abnormalities reminiscent of Alzheimer's disease. Western blot experiments further demonstrated a considerable increase in amyloid-β (Aβ) expression in the mouse brain following CSD. Meanwhile, the hub gene Prkcg was searched for in the cerebellum using RNA-seq and bioinformatics analysis. PKCγ (Prkcg) expression was significantly reduced, as demonstrated by RT-qPCR and Western blot validations. Additionally, CSD was associated with downregulated CREB expression, decreased expression of the endothelin-converting enzyme (ECE1), and increased phosphorylation of ERK1/2 downstream of PKCγ. These findings suggested that CSD down-regulated PKCγ expression, decreased ECE1 expression, impaired Aβ degradation, and affected the PKCγ/ERK/CREB pathway and the synthesis of memory-related proteins. Overall, this study highlighted how CSD modulated PKCγ-related metabolism, impacting Aβ clearance and the production of memory-related proteins. Such insights are crucial for understanding and preventing sporadic Alzheimer's disease (sAD) associated with CSD.
Collapse
Affiliation(s)
| | | | | | | | | | - Taijun Hang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 211100, China; (X.W.); (H.C.); (T.T.); (X.Z.); (S.Q.)
| | - Min Song
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 211100, China; (X.W.); (H.C.); (T.T.); (X.Z.); (S.Q.)
| |
Collapse
|
97
|
Bostick JW, Connerly TJ, Thron T, Needham BD, de Castro Fonseca M, Kaddurah-Daouk R, Knight R, Mazmanian SK. The microbiome shapes immunity in a sex-specific manner in mouse models of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.07.593011. [PMID: 38766238 PMCID: PMC11100721 DOI: 10.1101/2024.05.07.593011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
INTRODUCTION : Preclinical studies reveal that the microbiome broadly affects immune responses and deposition and/or clearance of amyloid-beta (Aβ) in mouse models of Alzheimer's disease (AD). Whether the microbiome shapes central and peripheral immune profiles in AD models remains unknown. METHODS : We examined adaptive immune responses in two mouse models containing AD- related genetic predispositions (3xTg and 5xFAD) in the presence or absence of the microbiome. RESULTS : T and B cells were altered in brain-associated and systemic immune tissues between genetic models and wildtype mice, with earlier signs of immune activity in females. Systemic immune responses were modulated by the microbiome and differed by sex. Further, the absence of a microbiome in germ-free mice resulted in reduced cognitive deficits, primarily in females. DISCUSSION : These data reveal sexual dimorphism in early signs of immune activity and microbiome effects, and highlight an interesting interaction between sex and the microbiome in mouse models of AD.
Collapse
|
98
|
Wong SQ, Ouellette A, McNamara A, Tam RA, Alexandrov A, Nawrocik-Madrid A, Sanchez JJ, Ginsburg BC, Andrade AA, Lapierre LR. Spatial memory in Alzheimer's disease 5XFAD mice is enhanced by XPO1 inhibitor KPT-330. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.21.619493. [PMID: 39484542 PMCID: PMC11527021 DOI: 10.1101/2024.10.21.619493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
The proteostatic decline in Alzheimer's disease is well established and improvement in proteostasis could potentially delay cognitive impairment. One emerging entry point to modulate proteostasis is the regulation of nucleo-cytoplasmic partitioning of proteins across the nuclear pore via karyopherins. The nuclear exportin XPO1 is a key regulator of proteostasis by driving the assembly of ribosomes and by modulating the process of autophagy. We recently found that XPO1 inhibitor KPT-330 (Selinexor), an FDA approved drug against multiple myelomas, enhances proteostasis, leading to benefits in models of neurodegenerative diseases in C. elegans and Drosophila. Here, we find that KPT-330 increases autophagy in murine neuronal cells and improves spatial memory performance in a murine model of Alzheimer's disease (5XFAD). Unexpectedly, general amyloid deposition in several brain regions was significantly increased by KPT-330, but specific regions, especially the thalamus, displayed significantly lower deposition, suggesting that XPO1 inhibition has regional-specific effects on proteostasis and amyloid plaque formation. Altogether, we conclude that XPO1 inhibition can improve cognition via spatially-specific reductions in amyloid deposition.
Collapse
Affiliation(s)
- Shi Quan Wong
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting St., Providence, Rhode Island 02912, USA
| | - Adia Ouellette
- Centre de Médecine de Précision du Nouveau Brunswick, 27 rue Providence, Moncton, New Brunswick, E1C 8X3, Canada
- Département de chimie et biochimie, Université de Moncton, 18 avenue Antonine Maillet, Moncton, New Brunswick, E1A 3E9, Canada
| | - Avery McNamara
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting St., Providence, Rhode Island 02912, USA
| | - Rachel A. Tam
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting St., Providence, Rhode Island 02912, USA
| | - Alexander Alexandrov
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting St., Providence, Rhode Island 02912, USA
| | - Acacia Nawrocik-Madrid
- Nathan Shock Center San Antonio, Analytical Pharmacology and Drug Evaluation Core, University of Texas in San Antonio, San Antonio, TX, USA
| | - Jesus J. Sanchez
- Nathan Shock Center San Antonio, Analytical Pharmacology and Drug Evaluation Core, University of Texas in San Antonio, San Antonio, TX, USA
| | - Brett C. Ginsburg
- Nathan Shock Center San Antonio, Analytical Pharmacology and Drug Evaluation Core, University of Texas in San Antonio, San Antonio, TX, USA
| | - Arturo A. Andrade
- Department of Neuroscience, Brown University, 185 Meeting St., Providence, Rhode Island 02912, USA
- Carney Institute for Brain Science, Brown University, 164 Angell St. Providence, Rhode Island. 02912, USA
| | - Louis R. Lapierre
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting St., Providence, Rhode Island 02912, USA
- Centre de Médecine de Précision du Nouveau Brunswick, 27 rue Providence, Moncton, New Brunswick, E1C 8X3, Canada
- Département de chimie et biochimie, Université de Moncton, 18 avenue Antonine Maillet, Moncton, New Brunswick, E1A 3E9, Canada
- Carney Institute for Brain Science, Brown University, 164 Angell St. Providence, Rhode Island. 02912, USA
| |
Collapse
|
99
|
Zhang Y, Chen X, Chen L, Shao M, Zhu W, Xing T, Guo T, Jia Q, Yang H, Yin P, Yan XX, Yu J, Li S, Li XJ, Yang S. Increased expression of mesencephalic astrocyte-derived neurotrophic factor (MANF) contributes to synapse loss in Alzheimer's disease. Mol Neurodegener 2024; 19:75. [PMID: 39425207 PMCID: PMC11490049 DOI: 10.1186/s13024-024-00771-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND The activation of endoplasmic reticulum (ER) stress is an early pathological hallmark of Alzheimer's disease (AD) brain, but how ER stress contributes to the onset and development of AD remains poorly characterized. Mesencephalic astrocyte-derived neurotrophic factor (MANF) is a non-canonical neurotrophic factor and an ER stress inducible protein. Previous studies reported that MANF is increased in the brains of both pre-symptomatic and symptomatic AD patients, but the consequence of the early rise in MANF protein is unknown. METHODS We examined the expression of MANF in the brain of AD mouse models at different pathological stages. Through behavioral, electrophysiological, and neuropathological analyses, we assessed the level of synaptic dysfunctions in the MANF transgenic mouse model which overexpresses MANF in the brain and in wild type (WT) mice with MANF overexpression in the hippocampus. Using proteomic and transcriptomic screening, we identified and validated the molecular mechanism underlying the effects of MANF on synaptic function. RESULTS We found that increased expression of MANF correlates with synapse loss in the hippocampus of AD mice. The ectopic expression of MANF in mice via transgenic or viral approaches causes synapse loss and defects in learning and memory. We also identified that MANF interacts with ELAV like RNA-binding protein 2 (ELAVL2) and affects its binding to RNA transcripts that are involved in synaptic functions. Increasing or decreasing MANF expression in the hippocampus of AD mice exacerbates or ameliorates the behavioral deficits and synaptic pathology, respectively. CONCLUSIONS Our study established MANF as a mechanistic link between ER stress and synapse loss in AD and hinted at MANF as a therapeutic target in AD treatment.
Collapse
Affiliation(s)
- Yiran Zhang
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Xiusheng Chen
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Laiqiang Chen
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Mingting Shao
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Wenzhen Zhu
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Tingting Xing
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Tingting Guo
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Qingqing Jia
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Huiming Yang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-Sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, Guangzhou, China
| | - Peng Yin
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Xiao-Xin Yan
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, China
| | - Jiandong Yu
- Department of Neurosurgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Shihua Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Su Yang
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China.
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China.
| |
Collapse
|
100
|
Ardanaz CG, de la Cruz A, Minhas PS, Hernández-Martín N, Pozo MÁ, Valdecantos MP, Valverde ÁM, Villa-Valverde P, Elizalde-Horcada M, Puerta E, Ramírez MJ, Ortega JE, Urbiola A, Ederra C, Ariz M, Ortiz-de-Solórzano C, Fernández-Irigoyen J, Santamaría E, Karsenty G, Brüning JC, Solas M. Astrocytic GLUT1 reduction paradoxically improves central and peripheral glucose homeostasis. SCIENCE ADVANCES 2024; 10:eadp1115. [PMID: 39423276 PMCID: PMC11488540 DOI: 10.1126/sciadv.adp1115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 09/13/2024] [Indexed: 10/21/2024]
Abstract
Astrocytes are considered an essential source of blood-borne glucose or its metabolites to neurons. Nonetheless, the necessity of the main astrocyte glucose transporter, i.e., GLUT1, for brain glucose metabolism has not been defined. Unexpectedly, we found that brain glucose metabolism was paradoxically augmented in mice with astrocytic GLUT1 reduction (GLUT1ΔGFAP mice). These mice also exhibited improved peripheral glucose metabolism especially in obesity, rendering them metabolically healthier. Mechanistically, we observed that GLUT1-deficient astrocytes exhibited increased insulin receptor-dependent ATP release, and that both astrocyte insulin signaling and brain purinergic signaling are essential for improved brain function and systemic glucose metabolism. Collectively, we demonstrate that astrocytic GLUT1 is central to the regulation of brain energetics, yet its depletion triggers a reprogramming of brain metabolism sufficient to sustain energy requirements, peripheral glucose homeostasis, and cognitive function.
Collapse
Affiliation(s)
- Carlos G. Ardanaz
- Department of Pharmaceutical Sciences, Division of Pharmacology, University of Navarra, 31008 Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Aida de la Cruz
- Laboratory of Local Translation in Neurons and Glia, Achucarro Basque Centre for Neuroscience, 48940 Leioa, Spain
| | - Paras S. Minhas
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nira Hernández-Martín
- Unidad de Cartografía Cerebral, Instituto Pluridisciplinar, Universidad Complutense de Madrid, 28040 Madrid, Spain
- PET Center, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Miguel Ángel Pozo
- Unidad de Cartografía Cerebral, Instituto Pluridisciplinar, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Unidad de Cartografía Cerebral, Instituto de Investigación Sanitaria, Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - M. Pilar Valdecantos
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM, Department of Metabolism and Cellular Signaling, Madrid 28029, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid 28029, Spain
- Universidad Francisco de Vitoria, Faculty of Experimental Sciences, Pozuelo de Alarcon, Madrid, Spain
| | - Ángela M. Valverde
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM, Department of Metabolism and Cellular Signaling, Madrid 28029, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid 28029, Spain
| | | | | | - Elena Puerta
- Department of Pharmaceutical Sciences, Division of Pharmacology, University of Navarra, 31008 Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - María J. Ramírez
- Department of Pharmaceutical Sciences, Division of Pharmacology, University of Navarra, 31008 Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Jorge E. Ortega
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
- Department of Pharmacology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Biobizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - Ainhoa Urbiola
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Imaging Platform, Foundation for Applied Medical Research (FIMA), University of Navarra (UNAV), 31008 Pamplona, Spain
| | - Cristina Ederra
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Imaging Platform, Foundation for Applied Medical Research (FIMA), University of Navarra (UNAV), 31008 Pamplona, Spain
| | - Mikel Ariz
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Imaging Platform, Foundation for Applied Medical Research (FIMA), University of Navarra (UNAV), 31008 Pamplona, Spain
- Department of Electrical, Electronic and Communications Engineering, Public University of Navarra, 31006 Pamplona, Spain
| | - Carlos Ortiz-de-Solórzano
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Imaging Platform, Foundation for Applied Medical Research (FIMA), University of Navarra (UNAV), 31008 Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Proteomics Platform, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain
| | - Gerard Karsenty
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, 701 West 168th Street, New York, NY, USA
| | - Jens C. Brüning
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, 50931 Cologne, Germany
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, 50924 Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
- National Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Maite Solas
- Department of Pharmaceutical Sciences, Division of Pharmacology, University of Navarra, 31008 Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| |
Collapse
|