1
|
Chen W, Johnston IN. Meta-analyses of executive function deficits in chemotherapy-treated rodent models. Neurosci Biobehav Rev 2025; 173:106131. [PMID: 40194612 DOI: 10.1016/j.neubiorev.2025.106131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/06/2025] [Accepted: 03/28/2025] [Indexed: 04/09/2025]
Abstract
People diagnosed with cancer who undergo chemotherapy commonly encounter cognitive changes, particularly in executive functions (EFs). EFs support goal-directed behaviours, with EF deficits implicated in various neurocognitive impairments. We conducted five meta-analyses of the rodent models to investigate the impact of chemotherapy across five EF domains. A systematic search across PubMed, Web of Science, Scopus, and PsycINFO yielded 56 eligible papers. Our findings supported the clinical literature suggesting the selective impact of chemotherapy on different EF domains. Specifically, chemotherapy-treated animals performed significantly more poorly than controls in tasks assessing working memory, behavioural flexibility, and problem-solving, with no significant group differences in inhibition or attention. Subgroup analyses revealed that alkylating agents, antitumor antibiotics, and combination therapies were strongly associated with working memory deficits, whereas mitotic inhibitors were not. Rodent species, strain, age, sex, number of treatments, and time of behavioural assessment since the end of treatment did not moderate the drug effect on any assessed EF domains. To increase the generalisability and translational validity of the results, the overall reporting quality of animal studies needs to be improved with more details on randomisation, blinding, sample sizes, and criteria for animal exclusions.
Collapse
Affiliation(s)
- Weiye Chen
- School of Psychology, University of Sydney, NSW 2006, Australia
| | - Ian N Johnston
- School of Psychology, University of Sydney, NSW 2006, Australia.
| |
Collapse
|
2
|
Yoshitome Queiroz L, Nunes Mariot L, Sousa Soares E, Urach Stahler C, Griebner G, de Moraes Machado G, M Gissoni J, Betta Canever J, Sordi R, Cimarosti H. Cecal slurry-induced sepsis in mice impairs cognition and decreases SUMO-2/3 conjugation. Behav Brain Res 2025; 485:115544. [PMID: 40118347 DOI: 10.1016/j.bbr.2025.115544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/07/2025] [Accepted: 03/15/2025] [Indexed: 03/23/2025]
Abstract
Sepsis is characterized by multiple organ dysfunction, dysregulation of the response to the infection process, and a high mortality rate in intensive care units. In addition, individuals who overcome sepsis often manifest cognitive deficits associated with neuroinflammation resulting from the entry of pro-inflammatory cytokines into the brain. Post-translational protein modifications, such as SUMOylation, can regulate the expression of pro-inflammatory genes during sepsis. Since SUMO-2/3 can play a role in pathological conditions, our aim was to investigate a potential link between sepsis-induced cognitive decline and SUMOylation by this isoform. Firstly, the cecal slurry model was induced by intraperitoneally injecting male Swiss mice with different volumes of a cecal solution. Following assessment of body temperature, mass and septic scores, the groups that received 300 μL and 350 μL of the cecal solution were selected for the behavioural tests, as they presented signs of sepsis without excessive mortality. Surviving animals were evaluated for cognition/memory and anxious/depressive-like behaviours through the open-field, object recognition, Y-maze, and tail suspension tests. Subsequently, SUMO-2/3 conjugation was determined in samples from the hippocampus and prefrontal cortex by Western blotting. Mice in the septic groups showed decreased locomotor activity, anxious-and depressive-like behaviours, as well as impaired memory. These deficits were accompanied by a decrease in SUMO-2/3 conjugation in the hippocampus and prefrontal cortex at 24 h and 10 days after the induction of the cecal slurry model. Taken together, our findings suggest that SUMOylation is impaired in septic animals and this could be related to the behavioural deficits seen in the surviving mice.
Collapse
Affiliation(s)
- Letícia Yoshitome Queiroz
- Postgraduate Program of Pharmacology Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil; Postgraduate Program of Neuroscience, Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil
| | - Luana Nunes Mariot
- Postgraduate Program of Pharmacology Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil
| | - Ericks Sousa Soares
- Postgraduate Program of Pharmacology Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil
| | - Carolina Urach Stahler
- Postgraduate Program of Pharmacology Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil
| | - Gustavo Griebner
- Postgraduate Program of Pharmacology Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil; Postgraduate Program of Neuroscience, Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil
| | - Gustavo de Moraes Machado
- Postgraduate Program of Neuroscience, Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil
| | - João M Gissoni
- Postgraduate Program of Pharmacology Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil
| | - Jaquelini Betta Canever
- Postgraduate Program of Neuroscience, Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil
| | - Regina Sordi
- Postgraduate Program of Pharmacology Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil
| | - Helena Cimarosti
- Postgraduate Program of Pharmacology Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil; Postgraduate Program of Neuroscience, Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil.
| |
Collapse
|
3
|
Rappeneau V, Tobinski AM, Guevara LMC, Meyer N, Jüngling K, Touma C. Role of the neuropeptide S receptor 1 rs324981 polymorphism in modulating emotionality and cognitive flexibility: Insights from a gene-edited mouse model. Behav Brain Res 2025; 485:115530. [PMID: 40064355 DOI: 10.1016/j.bbr.2025.115530] [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: 01/07/2025] [Revised: 02/25/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
The neuropeptide S (NPS) and its receptor (NPSR1) are involved in various physiological processes, including arousal, sleep, anxiety, memory, and stress responses in rodents. Recent attention has focused on the association between the NPS/NPSR1 system and stress-related disorders, particularly involving a specific single nucleotide polymorphism (SNP) in the NPSR1 gene (rs324981). This SNP causes an amino acid change at position 107 in the protein, reducing NPSR1 signalling potency; however, its effects on behavioural, cognitive, and physiological aspects relevant to stress-related disorders remain unclear. Addressing this topic, we characterized the behavioural phenotype of a gene-editing mouse model, expressing either the murine/ancestral NPSR1-I107 variant or the human NPSR1-N107 variant. Both, male and female mice underwent a comprehensive behavioural test battery assessing arousal, exploratory and anxiety-related behaviour under varying levels of novelty stress. Moreover, cognitive functions were evaluated with a special focus on cognitive flexibility using the Attentional Set Shifting Task (ASST). Additionally, markers of behavioural and endocrine stress reactivity were assessed as well as changes in body weight and body composition. Our results showed that NPSR1-N107 mice displayed increased anxiety-related behaviour compared to NPSR1-I107 mice, with no significant differences in arousal, exploratory behaviour or hormonal stress responses. However, NPSR1-N107 mice also exhibited better rule-reversal learning in the ASST, indicating enhanced cognitive flexibility. These findings provide clear evidence for a role of the NPSR1 rs324981 SNP in regulating emotionality and cognitive flexibility, underscoring the potential of the NPSR1-I107N mouse model for further elucidating the molecular mechanisms underlying stress-related disorders.
Collapse
Affiliation(s)
- Virginie Rappeneau
- Department of Behavioural Biology, Osnabrück University, Osnabrück, Germany.
| | - Ann-Marie Tobinski
- Department of Behavioural Biology, Osnabrück University, Osnabrück, Germany
| | | | - Neele Meyer
- Department of Behavioural Biology, Osnabrück University, Osnabrück, Germany
| | - Kay Jüngling
- Institute of Physiology I, University of Münster, Münster, Germany
| | - Chadi Touma
- Department of Behavioural Biology, Osnabrück University, Osnabrück, Germany.
| |
Collapse
|
4
|
Gupta S, Rishi V, Elipilla P, Aggarwal A. Upregulation of HDAC3 mediates behavioral impairment in the bile duct ligation model of hepatic encephalopathy. Int J Biol Macromol 2025; 307:141596. [PMID: 40054811 DOI: 10.1016/j.ijbiomac.2025.141596] [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: 08/08/2024] [Revised: 02/15/2025] [Accepted: 02/27/2025] [Indexed: 05/07/2025]
Abstract
Hepatic encephalopathy (HE), an outcome of chronic liver disease is characterized by behavioral impairments. The present study investigated the role of HDAC-mediated transcriptional regulation causing behavioral impairments in the bile duct ligation (BDL) model of HE. Post-BDL surgery in rats, dynamic alterations in liver function tests, liver morphology were observed. In BDL rats, histological staining in brain demonstrated reduced neuronal viability and warped neuronal architecture. Additionally, BDL animals showed impaired spatial learning, memory, and increased anxiety in the open field, Barnes maze, and Y maze tests. Further, the Golgi cox staining revealed a significantly altered spine density and spine clustering patterns of granular neuron in dentate gyrus of BDL rats. Concordantly, a significant downregulation of memory encoding genes was also observed in BDL rats that may account for aberrant behavior. Molecular analysis of modifiers, such as HDAC, showed significant changes in the expression of HDAC3 and HDAC6 in both the cortex and hippocampus of BDL rats. Upregulation of HDAC3 promoted its localization on the promoter of genes like c-Fos, NPAS4, Arc, and others, likely causing their decreased expression. Our findings suggest that increased HDAC3 activity downregulates key synaptic plasticity and memory-related genes, potentially driving neurobehavioral changes in BDL rats.
Collapse
Affiliation(s)
- Shiwangi Gupta
- National Agri-Food and Biomanufacturing Institute, Knowledge City, Sector-81, SAS Nagar, Punjab, India; Department of Biotechnology, Sector-25, BMS block I, Panjab University, Chandigarh, India
| | - Vikas Rishi
- National Agri-Food and Biomanufacturing Institute, Knowledge City, Sector-81, SAS Nagar, Punjab, India
| | - Pavani Elipilla
- National Agri-Food and Biomanufacturing Institute, Knowledge City, Sector-81, SAS Nagar, Punjab, India
| | - Aanchal Aggarwal
- National Agri-Food and Biomanufacturing Institute, Knowledge City, Sector-81, SAS Nagar, Punjab, India.
| |
Collapse
|
5
|
Zhang H, Xu Q, Jiang Z, Sun R, Wang Q, Liu S, Luan X, Campisi J, Kirkland JL, Zhang W, Sun Y. Targeting Senescence with Apigenin Improves Chemotherapeutic Efficacy and Ameliorates Age-Related Conditions in Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2412950. [PMID: 40265973 DOI: 10.1002/advs.202412950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 03/16/2025] [Indexed: 04/24/2025]
Abstract
Cellular senescence is a cell fate triggered by stressful stimuli and displays a hypersecretory feature, the senescence-associated secretory phenotype (SASP). Senescent cell burden increases with aging and contributes to age-related organ dysfunction and multiple chronic disorders. In this study, a large scale screening of a natural product library for senotherapeutic candidates is performed. Apigenin, a dietary flavonoid previously reported with antioxidant and anti-inflammatory activities, exhibits capacity for targeting senescent cells as a senomorphic agent. This compound blocks the interactions between ATM/p38MAPK and HSPA8, preventing the transition of an acute stress-associated phenotype (ASAP) toward the SASP. Mechanistically, apigenin targets peroxiredoxin 6 (PRDX6), an intracellular redox-active molecule, suppressing the iPLA2 activity of PRDX6 and disrupting downstream reactions underlying SASP development. Apigenin reduces the severity of cancer cell malignancy promoted by senescent stromal cells in culture, while restraining chemoresistance when combined with chemotherapy in anticancer regimens. In preclinical trials, apigenin improves the physical function of animals with a premature aging-like state, alleviating physical frailty and cognitive impairment. Together, the study demonstrates the feasibility of exploiting a natural compound with senomorphic capacity to achieve geroprotective effects by modulating the SASP, thus providing a baseline for future exploration of natural agents for alleviating age-related conditions.
Collapse
Affiliation(s)
- Hongwei Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P. R. China
| | - Qixia Xu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, P. R. China
| | - Zhirui Jiang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, P. R. China
| | - Rong Sun
- Department of Discovery Biology, Bioduro-Sundia, Zhangjiang Hi-Tech Park, Shanghai, 201210, P. R. China
| | - Qun Wang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P. R. China
| | - Sanhong Liu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P. R. China
| | - Xin Luan
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P. R. China
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - James L Kirkland
- Center for Advanced Gerotherapeutics, Cedars-Sinai Medical Center, Pacific Design Center, West Hollywood, CA, 90069, USA
- Division of Endocrinology, Diabetes and Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Weidong Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P. R. China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, P. R. China
| | - Yu Sun
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, P. R. China
- Department of Medicine and VAPSHCS, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
6
|
Li C, Yan Y, Stork O, Shen R, Behnisch T. The E3 Ubiquitin Ligase PRAJA1: A Key Regulator of Synaptic Dynamics and Memory Processes with Implications for Alzheimer's Disease. Int J Mol Sci 2025; 26:2909. [PMID: 40243483 PMCID: PMC11988436 DOI: 10.3390/ijms26072909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/13/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
The precise regulation of synaptic function by targeted protein degradation is fundamental to learning and memory, yet the roles of many brain-enriched E3 ubiquitin ligases in this process remain elusive. Here, we uncover a critical and previously unappreciated role for the E3 ubiquitin ligase PRAJA1 in orchestrating synaptic plasticity and hippocampus-dependent memory. Utilizing C57BL/6 and 5xFAD male mice and employing a multi-faceted approach including protein biochemistry, molecular biology, in vitro electrophysiology, and behavioral assays, we demonstrate that long-term potentiation (LTP) induction triggers a rapid, proteasome-dependent downregulation of PRAJA1 within the CA1 region of the hippocampus. Critically, selective knockdown of PRAJA1 in vivo profoundly enhanced both object recognition and spatial memory, while disrupting normal exploratory behavior. Mechanistically, we reveal that PRAJA1 acts as a key regulator of synaptic architecture and transmission: its downregulation leads to a reduction in key synaptic proteins and spine density, influencing the excitatory/inhibitory balance and facilitating synaptic plasticity. Conversely, increased PRAJA1 expression potentiates GABAergic transmission. Furthermore, we identify spinophilin as a novel substrate of PRAJA1, suggesting a direct molecular link between PRAJA1 and synaptic remodeling. Strikingly, our findings implicate dysregulation of PRAJA1 in the pathogenesis of Alzheimer's disease, positioning PRAJA1 as a potential therapeutic target for cognitive enhancement in neurodegenerative conditions. These results unveil PRAJA1 as a critical molecular brake on synaptic plasticity and memory formation, offering a promising new avenue for understanding and potentially treating memory impairment.
Collapse
Affiliation(s)
- Chuhan Li
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Yan Yan
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Oliver Stork
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany;
| | - Ruling Shen
- Shanghai Laboratory Animal Research Center, Shanghai 201203, China
| | - Thomas Behnisch
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| |
Collapse
|
7
|
Hoffman OR, Koehler JL, Espina JEC, Patterson AM, Gohar ES, Coleman EM, Schoenike BA, Espinosa-Garcia C, Paredes F, Varvel NH, Dingledine RJ, Maguire JL, Roopra AS. Disease modification upon 2 weeks of tofacitinib treatment in a mouse model of chronic epilepsy. Sci Transl Med 2025; 17:eadt0527. [PMID: 40106581 DOI: 10.1126/scitranslmed.adt0527] [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: 09/09/2024] [Accepted: 02/24/2025] [Indexed: 03/22/2025]
Abstract
All current drug treatments for epilepsy, a neurological disorder affecting more than 50 million people, merely treat symptoms, and a third of patients with epilepsy do not respond to medication. There are no disease-modifying treatments that may be administered briefly to patients to enduringly eliminate spontaneous seizures and reverse cognitive deficits. Applying network approaches to whole tissue and single-nucleus transcriptomic data collected from mouse models of temporal lobe epilepsy and publicly available transcriptomic data from human temporal lobectomy samples, we confirmed a previously described pattern of rapid and transient induction of the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway within days of epileptogenic insult. This was followed by a resurgent activation of the JAK/STAT pathway weeks to months later with the onset of spontaneous seizures. Targeting the first wave of JAK/STAT activation after epileptic insult did not prevent seizures. However, inhibition of the second wave with CP690550 (tofacitinib) over a 2-week period enduringly suppressed seizures, rescued deficits in spatial memory, and alleviated epilepsy-associated histopathological alterations. Seizure suppression lasted for at least 2 months after the final dose. These results indicate that reignition of inflammatory JAK/STAT3 signaling in chronic epilepsy opens a window for disease modification with the US Food and Drug Administration-approved, orally available drug CP690550.
Collapse
Affiliation(s)
- Olivia R Hoffman
- Department of Neuroscience, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
- Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Jennifer L Koehler
- Department of Neuroscience, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
- Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Jose Ezekiel Clemente Espina
- Department of Neuroscience, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Anna M Patterson
- Department of Neuroscience, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Emily S Gohar
- Department of Neuroscience, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Emanuel M Coleman
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Barry A Schoenike
- Department of Neuroscience, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Claudia Espinosa-Garcia
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Felipe Paredes
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Nicholas H Varvel
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Raymond J Dingledine
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jamie L Maguire
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Avtar S Roopra
- Department of Neuroscience, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
- Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
8
|
Rangel-Patiño CA, Mastachi-Loza CA, Carmen-Cristóbal JM, Ruiz-Gómez ML. Boldness and learning in an active foraging lizard. Behav Processes 2025; 226:105174. [PMID: 40049300 DOI: 10.1016/j.beproc.2025.105174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 02/12/2025] [Accepted: 03/03/2025] [Indexed: 03/18/2025]
Abstract
Foraging is a demanding activity for species that search intensively for food, and learning may help them minimise the costs associated with feeding. In different species, there is a relationship between personality (risk-taking) and learning, where bolder individuals learn fast and perform better in stable environments. On the other hand, shy individuals have slower learning rates because they spend more time paying attention to the environment, and their performance peaks under unstable environmental conditions. Therefore, we could expect that these differences will permeate other contexts, such as foraging mode. We evaluated learning and boldness in the active foraging lizard Aspidoscelis costatus costatus to establish if this association is present in reptiles. We found that males and females have similar learning abilities, with bolder individuals learning to find and consume food faster. Females were bolder than males in the presence of a novel object. We suggest that the results are related to the foraging ecology of the species, in which active foragers manifest risky behaviours (i.e. boldness, exploration, and higher activity) to search wide areas for prey, which may be enhanced by faster learning to reduce the costs associated with foraging.
Collapse
Affiliation(s)
- C A Rangel-Patiño
- Tecnológico Nacional de México, Tecnológico de Estudios Superiores de Huixquilucan, División de Biología, Laboratorio de Ecología y Conducta en Reptiles, México; Instituto Interamericano de Tecnología y Ciencias del Agua, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico.
| | - C A Mastachi-Loza
- Instituto Interamericano de Tecnología y Ciencias del Agua, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico.
| | - J M Carmen-Cristóbal
- Ecology and Behavior Lab, Facultad de Ciencias, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico.
| | - M L Ruiz-Gómez
- Ecology and Behavior Lab, Facultad de Ciencias, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico.
| |
Collapse
|
9
|
Depret N, Gleizes M, Moreau MM, Poirault-Chassac S, Quiedeville A, Carvalho SDS, Venugopal V, Abed ASA, Ezan J, Barthet G, Mulle C, Desmedt A, Marighetto A, Racca C, Montcouquiol M, Sans N. The correct connectivity of the DG-CA3 circuits involved in declarative memory processes depends on Vangl2-dependent planar cell polarity signaling. Prog Neurobiol 2025; 246:102728. [PMID: 39956311 DOI: 10.1016/j.pneurobio.2025.102728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/18/2025]
Abstract
In the hippocampus, dentate gyrus granule cells connect to CA3 pyramidal cells via their axons, the mossy fibers (Mf). The synaptic terminals of Mfs (Mf boutons, MfBs) form large and complex synapses with thorny excrescences (TE) on the proximal dendrites of CA3 pyramidal cells (PCs). MfB/TE synapses have distinctive "detonator" properties due to low initial release probability and large presynaptic facilitation. The molecular mechanisms shaping the morpho-functional properties of MfB/TE synapses are still poorly understood, though alterations in their morphology are associated with Down syndrome, intellectual disabilities, and Alzheimer's disease. Here, we identify the core PCP gene Vangl2 as essential to the morphogenesis and function of MfB/TE synapses. Vangl2 colocalises with the presynaptic heparan sulfate proteoglycan glypican 4 (GPC4) to stabilise the postsynaptic orphan receptor GPR158. Embryonic loss of Vangl2 disrupts the morphology of MfBs and TEs, impairs ultrastructural and molecular organisation, resulting in defective synaptic transmission and plasticity. In adult, the early loss of Vangl2 results in a number of hippocampus-dependent memory deficits including characteristic flexibility of declarative memory, organisation and retention of working / everyday-like memory. These deficits also lead to abnormal generalisation of memories to salient cues and diminished ability to form detailed contextual memories. Together, these results establish Vangl2 as a key regulator of DG-CA3 connectivity and functions.
Collapse
Affiliation(s)
- Noémie Depret
- Univ. Bordeaux, Inserm, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Marie Gleizes
- Univ. Bordeaux, Inserm, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Maïté Marie Moreau
- Univ. Bordeaux, Inserm, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | | | - Anne Quiedeville
- Univ. Bordeaux, Inserm, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | | | - Vasika Venugopal
- Univ. Bordeaux, Inserm, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Alice Shaam Al Abed
- Univ. Bordeaux, Inserm, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Jérôme Ezan
- Univ. Bordeaux, Inserm, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Gael Barthet
- Univ. Bordeaux, CNRS, IINS, UMR 5297, Bordeaux F-33000, France
| | | | - Aline Desmedt
- Univ. Bordeaux, Inserm, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Aline Marighetto
- Univ. Bordeaux, Inserm, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Claudia Racca
- Biosciences Institute, The Medical School, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Nathalie Sans
- Univ. Bordeaux, Inserm, Neurocentre Magendie, U1215, Bordeaux F-33000, France.
| |
Collapse
|
10
|
Carrel A, Napoli E, Hixson K, Carlsen J, Cruz Del Angel Y, Strode D, Busquet N, Kumar V, Wempe MF, Russek SJ, Brooks-Kayal AR. Ruxolitinib-dependent reduction of seizure load and duration is accompanied by spatial memory improvement in the rat pilocarpine model of temporal lobe epilepsy. Neurotherapeutics 2025; 22:e00506. [PMID: 39643584 PMCID: PMC12014301 DOI: 10.1016/j.neurot.2024.e00506] [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: 08/28/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024] Open
Abstract
Molecules with optimized pharmacokinetic properties selectively aimed at the inhibition of STAT3 phosphorylation in brain have recently emerged as potential disease modifying therapies for epilepsy. In the current study, pharmacological inhibition of JAK1/2 with the orally available, FDA-approved drug ruxolitinib, produced nearly complete inhibition of hippocampal STAT3 phosphorylation, and reduced the expression of its downstream target Cyclin D1, when administered to rats 30 min and 3 h after onset of pilocarpine-induced status epilepticus (SE). This effect was accompanied by significantly shorter seizure duration and lower overall seizure frequency throughout the 4 weeks of EEG recording, but did not completely prevent the development of epilepsy in ruxolitinib-treated male rats. Compared to DMSO-treated animals, administration of ruxolitinib also improved memory (Y maze) but did not impact motor function (open field) following SE. Taken together with our previous findings, the results of this study provide further evidence that inhibition of the JAK/STAT pathway may be a promising disease modifying strategy to reduce severity of acquired epilepsy after brain injury, but also point to the need to better understand and optimize inhibitors of this pathway.
Collapse
Affiliation(s)
- Andrew Carrel
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Eleonora Napoli
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Kathryn Hixson
- Graduate Program for Neuroscience, Center for Systems Neuroscience, Boston University, Boston, MA, USA
| | - Jessica Carlsen
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Yasmin Cruz Del Angel
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Dana Strode
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Nicolas Busquet
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Vijay Kumar
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz, Aurora, CO, USA
| | - Michael F Wempe
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz, Aurora, CO, USA; Department of Chemistry, Kentucky State University, Frankfort, KY, USA
| | - Shelley J Russek
- Graduate Program for Neuroscience, Center for Systems Neuroscience, Boston University, Boston, MA, USA; Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Amy R Brooks-Kayal
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA.
| |
Collapse
|
11
|
Wang H, Li Y, Li X, Sun Z, Yu F, Pashang A, Kulasiri D, Li HW, Chen H, Hou H, Zhang Y. The Primary Cilia are Associated with the Axon Initial Segment in Neurons. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407405. [PMID: 39804991 PMCID: PMC11884599 DOI: 10.1002/advs.202407405] [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: 07/02/2024] [Revised: 12/16/2024] [Indexed: 01/16/2025]
Abstract
The primary cilia serve as pivotal mediators of environmental signals and play crucial roles in neuronal responses. Disruption of ciliary function has been implicated in neuronal circuit disorders and aberrant neuronal excitability. However, the precise mechanisms remain elusive. To study the link between the primary cilia and neuronal excitability, manipulation of somatostatin receptor 3 (SSTR3) is investigated, as an example of how alterations in ciliary signaling may affect neuronal activity. It is found that aberrant SSTR3 expression perturbed not only ciliary morphology but also disrupted ciliary signaling cascades. Genetic deletion of SSTR3 resulted in perturbed spatial memory and synaptic plasticity. The axon initial segment (AIS) is a specialized region in the axon where action potentials are initiated. Interestingly, loss of ciliary SSTR3 led to decrease of Akt-dependent cyclic AMP-response element binding protein (CREB)-mediated transcription at the AIS, specifically downregulating AIS master organizer adaptor protein ankyrin G (AnkG) expression. In addition, alterations of other ciliary proteins serotonin 6 receptor (5-HT6R)and intraflagellar transport protein 88 (IFT88) also induced length changes of the AIS. The findings elucidate a specific interaction between the primary cilia and AIS, providing insight into the impact of the primary cilia on neuronal excitability and circuit integrity.
Collapse
Affiliation(s)
- Han Wang
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijing100871China
| | - Yu Li
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijing100871China
| | - Xin Li
- Beijing Life Science AcademyBeijing102200China
| | - Zehui Sun
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijing100871China
| | - Fengdan Yu
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijing100871China
| | - Abolghasem Pashang
- Centre for Advanced Computational Solutions (C‐fACS)AGLS facultyLincoln UniversityCanterbury7647New Zealand
| | - Don Kulasiri
- Centre for Advanced Computational Solutions (C‐fACS)AGLS facultyLincoln UniversityCanterbury7647New Zealand
| | - Hung Wing Li
- Department of ChemistryThe Chinese University of Hong KongHong Kong999077China
| | - Huan Chen
- Beijing Life Science AcademyBeijing102200China
| | - Hongwei Hou
- Beijing Life Science AcademyBeijing102200China
| | - Yan Zhang
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijing100871China
| |
Collapse
|
12
|
Padhy DS, Aggarwal P, Velayutham R, Banerjee S. Aerobic exercise and metformin attenuate the cognitive impairment in an experimental model of type 2 diabetes mellitus: focus on neuroinflammation and adult hippocampal neurogenesis. Metab Brain Dis 2025; 40:92. [PMID: 39775196 DOI: 10.1007/s11011-024-01489-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is a chronic metabolic disorder that increases the prevalence of cognitive impairment in the geriatric population. Aerobic exercise is an excellent non-pharmacological therapeutic strategy to prevent Alzheimer's disease, the most common form of dementia. The exact molecular mechanism of aerobic exercise (Exe) as an intervention to counter cognitive decline is far from clear. Metformin is a first-line agent against T2DM with neuroprotective properties. The present study assessed the role of treadmill exercise in combination with a low dose of metformin (Met; 70 mg/kg) in cognitive impairment and its associated molecular mechanism in T2DM rats. The experimental model of T2DM-associated cognitive decline was created by administration of a high-fat diet (HFD) with a low dose of streptozotocin (STZ; 35 mg/kg). Neurobehavioral assessments were performed to evaluate spatial recognition and fear-conditioned memory across the groups: control, HFD + STZ, HFD + STZ + Exe, and HFD + STZ + Exe + Met. In addition, we performed immunohistochemistry and western blotting on the rat hippocampal tissue from the above groups for protein expression studies. T2DM rats showed a significant cognitive decline compared to the control group, which improved in the long-term exercise and metformin co-administered animals. The level of neuroinflammation was significantly elevated in the hippocampal tissue of T2DM rats compared to the control and lowered after exercise and metformin treatment. T2DM reduced mature neurons and neurogenesis while increasing astrogliosis and microgliosis, ameliorated by exercise and metformin treatment. Moreover, T2DM impaired hippocampal neurogenesis by reducing the canonical Wnt/β-catenin pathway, which got upregulated in exercise and metformin-co-administered rats. Long-term aerobic exercise with metformin treatment ameliorated neuroinflammation and promoted adult hippocampal neurogenesis via upregulating the canonical Wnt/β-catenin pathway in T2DM rats.
Collapse
Affiliation(s)
- Dibya Sundar Padhy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)- Kolkata, Kolkata, West Bengal, 700054, India
| | - Punita Aggarwal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)- Kolkata, Kolkata, West Bengal, 700054, India
| | - Ravichandiran Velayutham
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER)- Kolkata, Kolkata, West Bengal, 700054, India.
| | - Sugato Banerjee
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)- Kolkata, Kolkata, West Bengal, 700054, India.
| |
Collapse
|
13
|
Liu Q, Sun Y, He B, Chen H, Wang L, Wang G, Zhang K, Zhao X, Zhang X, Shen D, Zhang X, Cui L. Gain-of-function ANXA11 mutation cause late-onset ALS with aberrant protein aggregation, neuroinflammation and autophagy impairment. Acta Neuropathol Commun 2025; 13:2. [PMID: 39755715 DOI: 10.1186/s40478-024-01919-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 12/20/2024] [Indexed: 01/06/2025] Open
Abstract
Mutations in the ANXA11 gene, encoding an RNA-binding protein, have been implicated in the pathogenesis of amyotrophic lateral sclerosis (ALS), but the underlying in vivo mechanisms remain unclear. This study examines the clinical features of ALS patients harboring the ANXA11 hotspot mutation p.P36R, characterized by late-onset motor neuron disease and occasional multi-system involvement. To elucidate the pathogenesis, we developed a knock-in mouse model carrying the p.P36R mutation. In both heterozygous and homozygous mutant mice, ANXA11 protein levels were comparable to those in wild-type. Both groups exhibited late-onset motor dysfunction at approximately 10 months of age, with similar survival rates to wild-type (> 24 months) and no signs of dementia. Pathological analysis revealed early abnormal aggregates in spinal cord motor neurons, cortical neurons, and muscle cells of homozygous mice. From 2 months of age, we observed mislocalized ANXA11 aggregates, SQSTM1/p62-positive inclusions, and cytoplasmic TDP-43 mislocalization, which intensified with disease progression. Importantly, mutant ANXA11 co-aggregated with TDP-43 and SQSTM1/p62-positive inclusions. Electron microscopy of the gastrocnemius muscle uncovered myofibrillar abnormalities, including sarcomeric disorganization, Z-disc dissolution, and subsarcolemmal electron-dense structures within autophagic vacuoles. Autophagic flux, initially intact at 2 months, was impaired by 9 months, as evidenced by decreased Beclin-1 and LC3BII/I levels and increased SQSTM1/p62 expression, coinciding with mTORC1 hyperactivation. Significant motor neuron loss and neuroinflammation were detected by 9 months, with marked muscle dystrophy apparent by 12 months compared to wild-type controls. These findings implicate the gain-of-function ANXA11 mutation drives late-onset motor neuron disease by early presymptomatic proteinopathy, progressive neuronal degeneration, neuroinflammation, and autophagic dysfunction.
Collapse
Affiliation(s)
- Qing Liu
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College (PUMC) and Chinese Academy of Medical Science (CAMS), Beijing, China.
| | - Ye Sun
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College (PUMC) and Chinese Academy of Medical Science (CAMS), Beijing, China
| | - Baodong He
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College (PUMC) and Chinese Academy of Medical Science (CAMS), Beijing, China
| | - Haodong Chen
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College (PUMC) and Chinese Academy of Medical Science (CAMS), Beijing, China
| | - Lijing Wang
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College (PUMC) and Chinese Academy of Medical Science (CAMS), Beijing, China
| | - Gaojie Wang
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College (PUMC) and Chinese Academy of Medical Science (CAMS), Beijing, China
| | - Kang Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Ximeng Zhao
- State Key Laboratory of Medical Molecular Biology, Mckusick-Zhang Center for Genetic Medicine, Institute of Basic Medical Sciences, PUMC and CAMS, Beijing, China
| | - Xinzhe Zhang
- State Key Laboratory of Medical Molecular Biology, Mckusick-Zhang Center for Genetic Medicine, Institute of Basic Medical Sciences, PUMC and CAMS, Beijing, China
| | - Dongchao Shen
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College (PUMC) and Chinese Academy of Medical Science (CAMS), Beijing, China
| | - Xue Zhang
- State Key Laboratory of Medical Molecular Biology, Mckusick-Zhang Center for Genetic Medicine, Institute of Basic Medical Sciences, PUMC and CAMS, Beijing, China.
- State Key Laboratory of Complex, Severe, and Rare Diseases, PUMCH, Beijing, China.
| | - Liying Cui
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College (PUMC) and Chinese Academy of Medical Science (CAMS), Beijing, China.
| |
Collapse
|
14
|
Soares ES, Queiroz LY, Canever JB, Griebner G, Stahler CU, Mansur DS, Prediger RDS, Cimarosti HI. SENP3 knockdown improves motor and cognitive impairments in the intranasal MPTP rodent model of Parkinson's disease. Physiol Behav 2025; 288:114725. [PMID: 39488250 DOI: 10.1016/j.physbeh.2024.114725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/04/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
Several mechanisms underlying Parkinson's disease (PD) remain unclear, and effective treatments are still lacking. The conjugation of the small ubiquitin-like modifier (SUMO), known as SUMOylation, to key proteins in PD has shown potential beneficial effects. Considering that this process is reversed by SUMO-specific proteases (SENPs), this study addressed the effects of increased SUMO-2/3 conjugation, mediated by SENP3 knockdown, in the intranasal 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) rodent model of PD. Two weeks after infusion of the shRNA-containing lentiviral vector into the dorsolateral striatum and one week following intranasal MPTP administration, male Wistar rats were evaluated using cognitive and motor behavioural tests. Infection efficiency was confirmed by detecting GFP expression in the dorsolateral striatum. SENP3 knockdown, verified by Western blotting, resulted in increased SUMO-2/3 conjugation. MPTP-administered rats displayed impairments in both recognition and spatial memories, while SENP3 knockdown prevented these deficits. Rats exposed to MPTP also exhibited motor dysfunction, which was ameliorated by SENP3 knockdown. These findings underscore the involvement of SUMO-2/3 conjugation in PD and its potential as a novel therapeutic target to counteract cognitive and motor impairments induced by neurodegeneration.
Collapse
Affiliation(s)
- Ericks S Soares
- Department of Pharmacology, Postgraduate Program in Pharmacology, Centre for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Leticia Y Queiroz
- Department of Pharmacology, Postgraduate Program in Pharmacology, Centre for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil; Postgraduate Program in Neuroscience, Centre for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Jaquelini B Canever
- Postgraduate Program in Neuroscience, Centre for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Gustavo Griebner
- Department of Pharmacology, Postgraduate Program in Pharmacology, Centre for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Carolina U Stahler
- Department of Pharmacology, Postgraduate Program in Pharmacology, Centre for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Daniel S Mansur
- Department of Microbiology, Immunology, and Parasitology, Centre for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Rui Daniel S Prediger
- Department of Pharmacology, Postgraduate Program in Pharmacology, Centre for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil; Postgraduate Program in Neuroscience, Centre for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Helena I Cimarosti
- Department of Pharmacology, Postgraduate Program in Pharmacology, Centre for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil; Postgraduate Program in Neuroscience, Centre for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil.
| |
Collapse
|
15
|
Shaw GA, Wegener AJ, Neigh GN. Chronic corticosterone administration alters synaptic mitochondrial function within the hippocampus of C57Bl/6NTac mice. Physiol Behav 2024; 287:114681. [PMID: 39209050 PMCID: PMC12021453 DOI: 10.1016/j.physbeh.2024.114681] [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: 03/22/2024] [Revised: 08/02/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Chronic activation of the hypothalamic-pituitary-adrenal axis increases circulating corticosterone levels, causing a host of downstream behavioral, molecular, and metabolic changes. Here, we assess the effects of chronic exogenous CORT administration on changes in behavior and mitochondrial respiration in hippocampal synaptosomes of male and female mice. Adult male (n = 15) and female (n = 17) C57Bl/6NTac mice were given 35ug/mL CORT or vehicle dissolved in their drinking water for 21 consecutive days. Chronic CORT increased piloerection in males only. Although volume of CORT-containing water consumed was similar between males and females, circulating plasma and fecal corticosterone levels were only elevated in CORT-exposed males. Behavioral effects of CORT were evident in the Y-maze such that CORT caused a decrease in direct revisits in both sexes. There was no observed presentation of anxiety-like behavior following chronic CORT administration. Functional hippocampal synaptosomes were analyzed for mitochondrial respiration using Agilent's Cell Mito Stress test. Chronic CORT caused a decrease in synaptic mitochondria basal respiration, maximal respiration, proton leak, and ATP production in both sexes. Despite only observing an effect of chronic CORT on corticosterone concentrations in fecal and blood samples of males, chronic CORT induced marked changes in hippocampal synaptic mitochondrial function of both sexes. These data highlight the importance of considering effects of stress hormone exposure on neural function even in the absence of measurable peripheral elevations in females.
Collapse
Affiliation(s)
- Gladys A Shaw
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - Amy J Wegener
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - Gretchen N Neigh
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
16
|
Alexandre-Silva V, Soares-Silva B, Pereira GC, Custódio-Silva AC, Carvalhinho-Lopes PS, Taliano LO, Lambertucci RH, Cavalcante MD, de Souza Araújo AA, Quintans-Júnior L, Dos Santos JR, Ribeiro AM. Eplingiella fruticosa leaf essential oil complexed with β-cyclodextrin exerts a neuroprotective effect in an Alzheimer's disease animal model induced by Streptozotocin. Metab Brain Dis 2024; 40:40. [PMID: 39579243 DOI: 10.1007/s11011-024-01484-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 11/13/2024] [Indexed: 11/25/2024]
Abstract
Alzheimer's Disease (AD) is physiopathologically marked by an accumulation of beta-amyloid peptide (Aβ), hyperphosphorylation of tau protein, inflammation, and oxidative stress in the brain tissue. While new drugs for AD have been approved, novel treatments are still needed. Eplingiella fruticosa (EF) has demonstrated anti-inflammatory and antioxidant properties, which may be beneficial against AD. This study aimed to evaluate the effects of EF leaf essential oil complexed with β-cyclodextrin in a sporadic AD model induced by streptozotocin (STZ). Male Wistar rats (5-6 months old) received an intracerebroventricular STZ injection (3 mg/kg) or vehicle, and were orally treated with vehicle, EF (5 mg/kg), or donepezil (5 mg/kg) for 14 days. Behavioral tests included olfactory discrimination, open field, novel object recognition, sucrose preference, and spontaneous alternation. Upon completion, rats were euthanatized, and their brains were analyzed for Aβ, tau, and IL-1β via immunohistochemistry, and for oxidative stress markers. STZ-treated rats showed memory deficits and anhedonia, accompanied by increased Aβ, tau, and IL-1β immunoreactivity in the olfactory bulb, cortex, hippocampus, and increased TBARS levels in the hippocampus. On the other hand, EF treatment improved short-term and working memory (p < 0.001), and reduced depressive-like behavior (p = 0.02). Additionally, EF treatment decreased Aβ, tau, and IL-1β immunoreactivity in the olfactory bulb, hippocampus and cortex (p < 0.05), and reduced TBARS levels (p = 0.04) and total oxidant status in the hippocampus (p = 0.03), and increased total antioxidant status in the cortex (p = 0.04). These findings suggest EF has neuroprotective effects against STZ-induced damage, indicating its potential as a novel compound for AD treatment.
Collapse
Affiliation(s)
- Vanessa Alexandre-Silva
- Department of Biosciences, Federal University of São Paulo, Santos, São Paulo, Brazil
- Department of Gerontology, Federal University of São Carlos, São Carlos, São Paulo, Brazil
| | - Beatriz Soares-Silva
- Department of Biosciences, Federal University of São Paulo, Santos, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Ma K, An C, Li M, Zhang Y, Ren M, Wei Y, Xu W, Wang R, Bai Y, Zhang H, Liu X, Ji S, Chen X, Zhu K. Dexmedetomidine Attenuated Neuron Death, Cognitive Decline, and Anxiety-Like Behavior by Inhibiting CXCL2 in CA1 Region of AD Mice. Drug Des Devel Ther 2024; 18:5351-5365. [PMID: 39605963 PMCID: PMC11600949 DOI: 10.2147/dddt.s489860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/16/2024] [Indexed: 11/29/2024] Open
Abstract
Purpose β-amyloid overload-induced neuroinflammation and neuronal loss are key pathological changes that occur during the progression of Alzheimer's disease (AD). Dexmedetomidine (Dex) exhibits neuroprotective and anti-inflammatory effects on the nervous system. However, the effect of Dex in AD mice remains unclear, and its neuroprotective regulatory mechanism requires further investigation. This study aimed to reveal how Dex protects against Aβ induced neuropathological changes and behavior dysfunction in AD mice. Methods An AD mouse model was established by the injection of Aβ into the brains of mice, followed by intraperitoneal injection with Dex. CXCL2 overexpression and Yohimbine, a Dex inhibitor, were used to investigate the role of Dex and CXCL2 in the regulation of neuronal loss, cognitive decline, and anxiety-like behavior in AD mice. Behavioral tests were performed to evaluate the cognitive and anxiety status of the mice. Nissl staining and immunofluorescence experiments were conducted to evaluate the status of the hippocampal neurons and astrocytes. qRT-PCR was performed to detect the expression of CXCL2, IL-1β, INOS, SPHK1, Bcl2, IFN-γ, and Caspase 1. The malondialdehyde (MDA) level was detected using an ELISA kit. Terminal TUNEL and Fluoro-Jade C (FJC) staining were used to measure the cell apoptosis rate. Results In AD mice, cognitive decline and anxiety-like behaviors were significantly improved by the Dex treatment. The number of neurons was increased in mice in the Dex + AD group compared to those in the AD group, and the number of astrocytes was not significantly different between the two groups. CXCL2, IL-1β, iNOS, and SPHK1 levels were significantly lower in Dex-treated AD mice than those in AD mice. Overloading of CXCL2 or Yohimbine reversed the protective effect of Dex on neuron number and cognitive and anxiety symptoms in AD mice. Conclusion Our results suggest that Dex exerts neuroprotective effects by downregulating CXCL2. Dex shows potential as a therapeutic drug for AD.
Collapse
Affiliation(s)
- Kaige Ma
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, 710061, People’s Republic of China
| | - Chanyuan An
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, 710061, People’s Republic of China
| | - Mai Li
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, 710061, People’s Republic of China
| | - Yuming Zhang
- Department of Anesthesiology, Shaanxi Provincial People’s Hospital, Xi’an, 710068, People’s Republic of China
| | - Minghe Ren
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, 710061, People’s Republic of China
| | - Yuyang Wei
- School of Forensic Medicine, Southern Medical University, Guangdong, 510515, People’s Republic of China
| | - Wenting Xu
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, 710061, People’s Republic of China
| | - Ruoxi Wang
- Department of Optometry, Fenyang College Shanxi Medical University, Fenyang, 032200, People’s Republic of China
| | - Yudan Bai
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, 710061, People’s Republic of China
| | - Hanyue Zhang
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, 710061, People’s Republic of China
| | - Xiyue Liu
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, 710061, People’s Republic of China
| | - Shengfeng Ji
- Department of Optometry, Fenyang College Shanxi Medical University, Fenyang, 032200, People’s Republic of China
| | - Xinlin Chen
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, 710061, People’s Republic of China
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, 710061, People’s Republic of China
| | - Kun Zhu
- Department of Neurology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
| |
Collapse
|
18
|
Amaro-Leal Â, Afonso AI, Machado F, Shvachiy L, Rocha I, Outeiro TF, Geraldes V. Dose-Dependent Cognitive Decline, Anxiety, and Locomotor Impairments Induced by Doxorubicin: Evidence from an Animal Model. BIOLOGY 2024; 13:939. [PMID: 39596894 PMCID: PMC11592173 DOI: 10.3390/biology13110939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024]
Abstract
Cognitive impairment and anxiety are common side effects of chemotherapy, particularly with the use of doxorubicin (DOX), known as "chemobrain". This study aimed to examine the dose-dependent effects of DOX on cognitive decline, anxiety, and locomotor activity in healthy female Wistar rats. The rats were divided into groups receiving low (2 mg/kg), intermediate (4 mg/kg), and high (5 mg/kg) doses of DOX for four weeks, alongside a control group. Behavioral tests, including open field, elevated plus maze, and Y-maze tests, assessed anxiety, locomotion, and cognitive performance, while brain tissue analysis evaluated neuroinflammation using markers such as GFAP and Iba-1. The results showed that all doses of DOX induced anxiety-like behavior, reduced locomotion, and caused neuroinflammation in the hippocampus, with more severe effects at higher doses. Notably, high-dose DOX also caused short-term memory deficits. These findings highlight the dose-dependent nature of DOX's impact on behavior and cognition, suggesting that DOX plays a key role in the development of cognitive symptoms during chemotherapy. Further research is needed to understand the mechanisms behind these effects and to explore potential interventions.
Collapse
Affiliation(s)
- Ângela Amaro-Leal
- Egas Moniz Center for Interdisciplinary Research (CiiEM), Egas Moniz School of Health & Science, 2829-511 Almada, Portugal
| | - Ana I. Afonso
- Faculdade de Medicina, Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028 Lisbon, Portugal
- Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina, Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028 Lisbon, Portugal
| | - Filipa Machado
- Faculdade de Medicina, Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028 Lisbon, Portugal
| | - Liana Shvachiy
- Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina, Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028 Lisbon, Portugal
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Isabel Rocha
- Faculdade de Medicina, Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028 Lisbon, Portugal
- Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina, Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028 Lisbon, Portugal
| | - Tiago F. Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37073 Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, 37075 Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK
- Scientific Employee with an Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 37075 Göttingen, Germany
| | - Vera Geraldes
- Faculdade de Medicina, Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028 Lisbon, Portugal
- Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina, Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028 Lisbon, Portugal
| |
Collapse
|
19
|
Sabet FS, Dabirmanesh B, Sabet HS, Zarei P, Hosseini M, Fathollahi Y, Khajeh K. The electro-responsive nanoliposome as an on-demand drug delivery platform for epilepsy treatment. Int J Pharm 2024; 664:124610. [PMID: 39168285 DOI: 10.1016/j.ijpharm.2024.124610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/14/2024] [Accepted: 08/17/2024] [Indexed: 08/23/2024]
Abstract
Nano-based drug delivery systems are regarded as a promising tool for efficient epilepsy treatment and seizure medication with the least general side effects and socioeconomic challenges. In the current study, we have designed a smart nanoscale drug delivery platform and applied it in the kindling model of epilepsy that is triggered rapidly by epileptic discharges and releases anticonvulsant drugs in situ, such as carbamazepine (CBZ). The CBZ-loaded electroactive ferrocene nanoliposomes had an average diameter of 100.6 nm, a surface charge of -7.08 mV, and high drug encapsulation efficiency (85.4 %). A significant increase in liposome size was observed in response to direct current (50-500 μA) application. This liposome-based drug delivery system can release CBZ at a fast rate in response to both direct current and pulsatile electrical stimulation in vitro. The CBZ-liposome can release the anticonvulsant drug upon epileptiform activity in the kindled rat model and can decline electrographic and behavioral seizure activity in response to electrical stimulation of the hippocampus with an initially subconvulsive current. With satisfactory biosafety results, this "smart" nanocarrier has promising potential as an effective and safe drug delivery system to improve the therapeutic index of antiepileptic drugs.
Collapse
Affiliation(s)
- Fereshte Sadat Sabet
- Department of Nanobiotechnology, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran.
| | - Bahareh Dabirmanesh
- Department of Biochemistry, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran.
| | - Hoorie Sadat Sabet
- Department of Life Science Engineering, Faculty of New Sciences & Technologies, University of Tehran, Tehran, Iran.
| | - Parisa Zarei
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Morteza Hosseini
- Department of Life Science Engineering, Faculty of New Sciences & Technologies, University of Tehran, Tehran, Iran.
| | - Yaghoub Fathollahi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Khosro Khajeh
- Department of Nanobiotechnology, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran; Department of Biochemistry, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
20
|
Reddy DS, Li Y, Qamari T, Ramakrishnan S. Behavioral Assays for Comprehensive Evaluation of Cognitive and Neuropsychiatric Comorbidities of Traumatic Brain Injury and Chronic Neurological Disorders. Curr Protoc 2024; 4:e70019. [PMID: 39422165 DOI: 10.1002/cpz1.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Neurological deficits, psychiatric disorders, and cognitive impairments often accompany stroke, brain injury, epilepsy, and many neurological disorders, which present intricate comorbidities that challenge recognition and management. There are many tools and paradigms for evaluating learning, memory, anxiety, and depression-like behaviors in lab animal models of brain disorders. However, there is a significant gap between clinical observations and experimental models, which limit understanding of the complex interplay between chronic brain conditions and their impact on cognitive dysfunction and psychiatric impairments. This article describes an overview of experimental rationale, methods, protocols, and strategies for evaluating sensorimotor, affective and cognitive-associated comorbid behaviors in epilepsy, traumatic brain injury (TBI), stroke, spinal cord injury (SCI), and many other neurological disorders. First, we delve into clinical evidence elucidating the profound impact of comorbidities, e.g., psychiatric disorders and cognitive deficits, in individuals with epilepsy. Then, we discuss diverse approaches to assess these comorbidities in experimental models of brain diseases. Finally, we explore the methodologies for assessing motor function, sensorimotor, behavior, and psychiatric health. We cover strategies and protocols enabling these assays, including implementing behavioral paradigms to assess learning and memory, anxiety, and depression-like behaviors in rodents in health and disease conditions. It is essential to consider a comprehensive battery of tests to investigate various behavioral deficits, considering environment, age, and sex differences relevant to the disease, such as TBI, SCI, epilepsy, stroke, and other complex neurological conditions. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas
- Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, Bryan, Texas
| | - Yue Li
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Taha Qamari
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Sreevidhya Ramakrishnan
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| |
Collapse
|
21
|
Rezqaoui A, Boumlah S, El Hessni A, El Brouzi MY, El Hamzaoui A, Ibouzine-Dine L, Benkirane S, Adnani M, Mesfioui A. Evaluating the Protective Effects of Melatonin Against Chronic Iron Administration in Male Wistar Rats: a Comparative Analysis of Affective, Cognitive, and Oxidative Stress with EDTA Chelator. Biol Trace Elem Res 2024; 202:4531-4546. [PMID: 38146034 DOI: 10.1007/s12011-023-04006-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/06/2023] [Indexed: 12/27/2023]
Abstract
Iron is the dominant metal in the brain and is distributed widely. However, it can lead to various neuropathological and neurobehavioral abnormalities as well as oxidative stress. On the other hand, melatonin, a pineal hormone, is known for its neuroprotective properties, as well as its ability to act as a natural chelator against oxidative stress. It has also been used as an antidepressant and anxiolytic. The study investigated the potential of melatonin and EDTA treatment to prevent anxiety, depressive behavior, and memory impairment in male rats induced by chronic iron administration, and its connection to oxidative stress regulation in the hippocampus and prefrontal cortex. The rats were divided into six groups and intraperitoneally injected for 8 weeks with NaCl solution (control), iron sulfate (1 mg/kg), melatonin (4 mg/kg), EDTA (4 mg/kg), 1 mg/kg of iron + 4 mg/kg of melatonin, or 1 mg/kg of iron + 4 mg/kg of EDTA. In this study, we performed a neurobehavioral assessment and biochemical determinations of oxidative stress levels in the hippocampus and prefrontal cortex of each animal. The results indicate that chronic exposure to iron sulfate induced anxiety-like depressive behavior, and cognitive impairment also increased the levels of lipid peroxidation and nitric oxide, and reduced the activity of catalase in the hippocampus and prefrontal cortex in male Wistar rats, suggesting the induction of oxidative stress. In contrast, these alterations were reversed by melatonin better than EDTA. The results of this study show that melatonin protects against the neurobehavioral changes caused by iron, which may be associated with decreasing oxidative stress in the hippocampus and prefrontal cortex.
Collapse
Affiliation(s)
- Ayoub Rezqaoui
- Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, B.P 242, Kenitra, Morocco.
| | - Soufiane Boumlah
- Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, B.P 242, Kenitra, Morocco
| | - Aboubaker El Hessni
- Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, B.P 242, Kenitra, Morocco
| | - Mohamed Yassine El Brouzi
- Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, B.P 242, Kenitra, Morocco
| | - Abdelghafour El Hamzaoui
- Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, B.P 242, Kenitra, Morocco
| | - Laila Ibouzine-Dine
- Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, B.P 242, Kenitra, Morocco
| | - Samir Benkirane
- Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, B.P 242, Kenitra, Morocco
| | - Manal Adnani
- Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, B.P 242, Kenitra, Morocco
| | - Abdelhalem Mesfioui
- Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, B.P 242, Kenitra, Morocco
| |
Collapse
|
22
|
Luna-Munguia H, Gasca-Martinez D, Garay-Cortes A, Coutiño D, Regalado M, de Los Rios E, Villaseñor P, Hidalgo-Flores F, Flores-Guapo K, Benito BY, Concha L. Selective Medial Septum Lesions in Healthy Rats Induce Longitudinal Changes in Microstructure of Limbic Regions, Behavioral Alterations, and Increased Susceptibility to Status Epilepticus. Mol Neurobiol 2024; 61:1-21. [PMID: 38443731 DOI: 10.1007/s12035-024-04069-9] [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: 09/25/2023] [Accepted: 02/26/2024] [Indexed: 03/07/2024]
Abstract
Septo-hippocampal pathway, crucial for physiological functions and involved in epilepsy. Clinical monitoring during epileptogenesis is complicated. We aim to evaluate tissue changes after lesioning the medial septum (MS) of normal rats and assess how the depletion of specific neuronal populations alters the animals' behavior and susceptibility to establishing a pilocarpine-induced status epilepticus. Male Sprague-Dawley rats were injected into the MS with vehicle or saporins (to deplete GABAergic or cholinergic neurons; n = 16 per group). Thirty-two animals were used for diffusion tensor imaging (DTI); scanned before surgery and 14 and 49 days post-injection. Fractional anisotropy and apparent diffusion coefficient were evaluated in the fimbria, dorsal hippocampus, ventral hippocampus, dorso-medial thalamus, and amygdala. Between scans 2 and 3, animals were submitted to diverse behavioral tasks. Stainings were used to analyze tissue alterations. Twenty-four different animals received pilocarpine to evaluate the latency and severity of the status epilepticus 2 weeks after surgery. Additionally, eight different animals were only used to evaluate the neuronal damage inflicted on the MS 1 week after the molecular surgery. Progressive changes in DTI parameters in both white and gray matter structures of the four evaluated groups were observed. Behaviorally, the GAT1-saporin injection impacted spatial memory formation, while 192-IgG-saporin triggered anxiety-like behaviors. Histologically, the GABAergic toxin also induced aberrant mossy fiber sprouting, tissue damage, and neuronal death. Regarding the pilocarpine-induced status epilepticus, this agent provoked an increased mortality rate. Selective septo-hippocampal modulation impacts the integrity of limbic regions crucial for certain behavioral skills and could represent a precursor for epilepsy development.
Collapse
Affiliation(s)
- Hiram Luna-Munguia
- Departamento de Neurobiologia Conductual y Cognitiva, Instituto de Neurobiologia, Universidad Nacional Autonoma de Mexico, Campus UNAM-Juriquilla, 76230, Queretaro, Mexico.
| | - Deisy Gasca-Martinez
- Departamento de Neurobiologia Conductual y Cognitiva, Instituto de Neurobiologia, Universidad Nacional Autonoma de Mexico, Campus UNAM-Juriquilla, 76230, Queretaro, Mexico
- Unidad de Analisis Conductual, Instituto de Neurobiologia, Universidad Nacional Autonoma de Mexico, Campus UNAM-Juriquilla, 76230, Queretaro, Mexico
| | - Alejandra Garay-Cortes
- Departamento de Neurobiologia Conductual y Cognitiva, Instituto de Neurobiologia, Universidad Nacional Autonoma de Mexico, Campus UNAM-Juriquilla, 76230, Queretaro, Mexico
| | - Daniela Coutiño
- Departamento de Neurobiologia Conductual y Cognitiva, Instituto de Neurobiologia, Universidad Nacional Autonoma de Mexico, Campus UNAM-Juriquilla, 76230, Queretaro, Mexico
| | - Mirelta Regalado
- Departamento de Neurobiologia Conductual y Cognitiva, Instituto de Neurobiologia, Universidad Nacional Autonoma de Mexico, Campus UNAM-Juriquilla, 76230, Queretaro, Mexico
| | - Ericka de Los Rios
- Departamento de Neurobiologia Conductual y Cognitiva, Instituto de Neurobiologia, Universidad Nacional Autonoma de Mexico, Campus UNAM-Juriquilla, 76230, Queretaro, Mexico
- Unidad de Microscopia, Instituto de Neurobiologia, Universidad Nacional Autonoma de Mexico, Campus UNAM-Juriquilla, 76230, Queretaro, Mexico
| | - Paulina Villaseñor
- Departamento de Neurobiologia Conductual y Cognitiva, Instituto de Neurobiologia, Universidad Nacional Autonoma de Mexico, Campus UNAM-Juriquilla, 76230, Queretaro, Mexico
| | - Fernando Hidalgo-Flores
- Departamento de Neurobiologia Conductual y Cognitiva, Instituto de Neurobiologia, Universidad Nacional Autonoma de Mexico, Campus UNAM-Juriquilla, 76230, Queretaro, Mexico
| | - Karen Flores-Guapo
- Departamento de Neurobiologia Conductual y Cognitiva, Instituto de Neurobiologia, Universidad Nacional Autonoma de Mexico, Campus UNAM-Juriquilla, 76230, Queretaro, Mexico
| | - Brandon Yair Benito
- Departamento de Neurobiologia Conductual y Cognitiva, Instituto de Neurobiologia, Universidad Nacional Autonoma de Mexico, Campus UNAM-Juriquilla, 76230, Queretaro, Mexico
| | - Luis Concha
- Departamento de Neurobiologia Conductual y Cognitiva, Instituto de Neurobiologia, Universidad Nacional Autonoma de Mexico, Campus UNAM-Juriquilla, 76230, Queretaro, Mexico
| |
Collapse
|
23
|
Abedi A, Foroutan T, Mohaghegh Shalmani L, Dargahi L. Sex-dependent susceptibility to brain metabolic dysfunction and memory impairment in response to pre and postnatal high-fat diet. J Nutr Biochem 2024; 132:109675. [PMID: 38945454 DOI: 10.1016/j.jnutbio.2024.109675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/15/2024] [Accepted: 05/31/2024] [Indexed: 07/02/2024]
Abstract
The developing brain is sensitive to the impacts of early-life nutritional intake. This study investigates whether maternal high fat diet (HFD) causes glucose metabolism impairment, neuroinflammation, and memory impairment in immature and adult offspring, and whether it may be affected by postweaning diets in a sex-dependent manner in adult offspring. After weaning, female rats were fed HFD (55.9% fat) or normal chow diet (NCD; 10% fat) for 8 weeks before mating, during pregnancy, and lactation. On postnatal day 21 (PND21), the male and female offspring of both groups were split into two new groups, and NCD or HFD feeding was maintained until PND180. On PND21 and PND180, brain glucose metabolism, inflammation, and Alzheimer's pathology-related markers were by qPCR. In adult offspring, peripheral insulin resistance parameters, spatial memory performance, and brain glucose metabolism (18F-FDG-PET scan and protein levels of IDE and GLUT3) were assessed. Histological analysis was also performed on PND21 and adult offspring. On PND21, we found that maternal HFD affected transcript levels of glucose metabolism markers in both sexes. In adult offspring, more profoundly in males, postweaning HFD in combination with maternal HFD induced peripheral and brain metabolic disturbances, impaired memory performance and elevated inflammation, dementia risk markers, and neuronal loss. Our results suggest that maternal HFD affects brain glucose metabolism in the early ages of both sexes. Postweaning HFD sex-dependently causes brain metabolic dysfunction and memory impairment in later-life offspring; effects that can be worsened in combination with maternal HFD.
Collapse
Affiliation(s)
- Azam Abedi
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Tahereh Foroutan
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| | - Leila Mohaghegh Shalmani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
24
|
Christensen A, McGill CJ, Qian W, Pike CJ. Effects of obesogenic diet and 17β-estradiol in female mice with APOE 3/3, 3/4, and 4/4 genotypes. Front Aging Neurosci 2024; 16:1415072. [PMID: 39347015 PMCID: PMC11427389 DOI: 10.3389/fnagi.2024.1415072] [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: 04/09/2024] [Accepted: 08/26/2024] [Indexed: 10/01/2024] Open
Abstract
The main genetic risk factor for Alzheimer's disease (AD) is the apolipoprotein E ε4 allele (APOE4). AD risk associated with APOE4 disproportionately affects women. Furthermore, human and rodent studies indicate that the cognitive deficits associated with APOE4 are greater in females. One modifiable AD risk factor is obesity during middle age. Given that approximately two-thirds of US adults are overweight, it is important to understand how obesity affects AD risk, how it interacts with APOE4, and the extent to which its detrimental effects can be mitigated with therapeutics. One intervention study for women is estrogen-based hormone therapy, which can exert numerous health benefits when administered in early middle age. No experimental studies have examined the interactions among APOE4, obesity, and hormone therapy in aging females. To begin to explore these issues, we considered how obesity outcomes are affected by treatment with estradiol at the onset of middle age in female mice with human APOE3 and APOE4. Furthermore, to explore how gene dosage affects outcomes, we compared mice homozygous for APOE3 (3/3) and homozygous (4/4) or hemizygous (3/4) for APOE4. Mice were examined over a 4-month period that spans the transition into reproductive senescence, a normal age-related change that models many aspects of human perimenopause. Beginning at 5 months of age, mice were maintained on a control diet (10% fat) or high-fat diet (HFD; 60% fat). After 8 weeks, by which time obesity was present in all HFD groups, mice were implanted with an estradiol or vehicle capsule that was maintained for the final 8 weeks. Animals were assessed on a range of metabolic and neural measures. Overall, APOE4 was associated with poorer metabolic function and cognitive performance. However, an obesogenic diet induced relatively greater impairments in metabolic function and cognitive performance in APOE3/3 mice. Estradiol treatment improved metabolic and cognitive outcomes across all HFD groups, with APOE4/4 generally exhibiting the greatest benefit. APOE3/4 mice were intermediate to the homozygous genotypes on many measures but also exhibited unique profiles. Together, these findings highlight the importance of the APOE genotype as a modulator of the risks associated with obesity and the beneficial outcomes of estradiol.
Collapse
Affiliation(s)
| | | | | | - Christian J. Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
25
|
Anarghou H, Malqui H, Ihbour S, Laaroussi M, Essaidi O, Fetoui H, Bouhrim M, Najimi M, Chigr F. Impact of glyphosate-based herbicide exposure through maternal milk on offspring's antioxidant status, neurodevelopment, and behavior. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6591-6609. [PMID: 38466353 DOI: 10.1007/s00210-024-03035-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 03/01/2024] [Indexed: 03/13/2024]
Abstract
Glyphosate-based Herbicide (GBH) is a widely used pesticide that functions as a broad-spectrum, non-selective herbicide. Despite advanced research to describe the neurotoxic potential of GBH, the harmful effects on maternal behavior and neurodevelopment of offspring remain unclear. This study was conducted to highlight the effects of GBH on the antioxidant system, anxiety traits, social interaction, and cognitive and sensorimotor functions in pups exposed to 25 or 50 mg/l daily via their mother's milk. Concerning the biochemical biomarkers, GBH administered during the early stages of development negatively affected the status of antioxidant enzymes and lipid peroxidation in the brain structures of the pups. Furthermore, our results showed a significant decrease in acetylcholinesterase (AChE) specific activity within the brains of treated pups. The results of the behavioral tests indicated that the treated offspring developed anxiety, memory, and sociability disorders, as evidenced by the Open Field, Y-maze, object recognition task, and social interaction tests. Through neurodevelopmental testing, we also showed sensorimotor impairment (righting reflex and negative geotaxis) and abnormal maternal behavior. Altogether, our study clearly demonstrates that the developing brain is sensitive to GBH.
Collapse
Affiliation(s)
- Hammou Anarghou
- Biological Engineering Laboratory, Faculty of Sciences and Techniques, Sultan Moulay Slimane University, Beni Mellal, Morocco.
- High Institute of Nursing Professions and Health Techniques Dakhla Annex, Dakhla, Morocco.
| | - Hafsa Malqui
- Biological Engineering Laboratory, Faculty of Sciences and Techniques, Sultan Moulay Slimane University, Beni Mellal, Morocco
| | - Said Ihbour
- Biological Engineering Laboratory, Faculty of Sciences and Techniques, Sultan Moulay Slimane University, Beni Mellal, Morocco
| | - Meriem Laaroussi
- Biological Engineering Laboratory, Faculty of Sciences and Techniques, Sultan Moulay Slimane University, Beni Mellal, Morocco
| | - Oumaima Essaidi
- Biological Engineering Laboratory, Faculty of Sciences and Techniques, Sultan Moulay Slimane University, Beni Mellal, Morocco
| | - Hamadi Fetoui
- Laboratory of Toxicology-Microbiology and Environmental Health (17ES06), Faculty of Sciences of Sfax, University of Sfax, BP1171, 3000, Sfax, Tunisia
| | - Mohamed Bouhrim
- Biological Engineering Laboratory, Faculty of Sciences and Techniques, Sultan Moulay Slimane University, Beni Mellal, Morocco
- Laboratories TBC, Laboratory of Pharmacology, Pharmacokinetics and Clinical Pharmacy, University of Lille, Faculty of Pharmacy, F-59000, Lille, France
| | - Mohamed Najimi
- Biological Engineering Laboratory, Faculty of Sciences and Techniques, Sultan Moulay Slimane University, Beni Mellal, Morocco
| | - Fatiha Chigr
- Biological Engineering Laboratory, Faculty of Sciences and Techniques, Sultan Moulay Slimane University, Beni Mellal, Morocco
| |
Collapse
|
26
|
Park W, Kim Y, Cho SH. Protective Effects and Mechanism of Heracleum moellendorffii Hance on Alcohol-Induced Cognitive Decline in Mice. Int J Mol Sci 2024; 25:8526. [PMID: 39126094 PMCID: PMC11313269 DOI: 10.3390/ijms25158526] [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] [Revised: 07/31/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
Chronic and continuous alcohol consumption increases the risk of cognitive decline and may lead to alcohol-related dementia. We investigated the potential of Heracleum moellendorffii Hance root extract (HME) for treating alcohol-related cognitive impairment. Behavioral tests evaluated the effects of HME on cognitive function and depression. Changes in hippocampus and liver tissues were evaluated by Western blotting and H&E staining. The group treated with HME 200 mg/kg showed a significant increase in spontaneous alternation in Y-maze and a decrease in immobility in a forced swimming test (FST) compared to the vehicle-treated group. These results suggest that HME can restore memory deficits and reverse depressive symptoms caused by chronic alcohol consumption. The HME-treated group also upregulated brain-derived neurotrophic factor (BDNF), phosphorylated extracellular signal-regulated kinase 1/2 (ERK1/2), and phosphorylated cAMP response element-binding protein (CREB) in the hippocampus. Additionally, it reduced lipid vacuolation in the liver and increased the expression of aldehyde dehydrogenase 1 (ADH1). The administration of HME improves cognitive impairment and reverses depressive symptoms due to alcohol consumption, restoring neural plasticity in the hippocampus and alcohol metabolism in the liver. These findings suggest that HME is a promising treatment for alcohol-related brain disorders. Molecular mechanisms underlying the therapeutic effects of HME and its active ingredients should be investigated further.
Collapse
Affiliation(s)
- Woohee Park
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Yunna Kim
- Department of Neuropsychiatry, College of Korean Medicine, Kyung Hee University Medical Center, Kyung Hee University, Seoul 02447, Republic of Korea;
- Research Group of Neuroscience, East-West Medical Research Institute, WHO Collaborating Center, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seung-Hun Cho
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
- Department of Neuropsychiatry, College of Korean Medicine, Kyung Hee University Medical Center, Kyung Hee University, Seoul 02447, Republic of Korea;
- Research Group of Neuroscience, East-West Medical Research Institute, WHO Collaborating Center, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
27
|
Chen XQ, Becker A, Albay R, Nguyen PD, Karachentsev D, Roberts AJ, Rynearson KD, Tanzi RE, Mobley WC. γ-Secretase Modulator BPN15606 Reduced Aβ42 and Aβ40 and Countered Alzheimer-Related Pathologies in a Mouse Model of Down Syndrome. Ann Neurol 2024; 96:390-404. [PMID: 38747498 PMCID: PMC11236496 DOI: 10.1002/ana.26958] [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: 11/28/2023] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/30/2024]
Abstract
OBJECTIVES Due to increased gene dose for the amyloid precursor protein (APP), elderly adults with Down syndrome (DS) are at a markedly increased risk of Alzheimer's disease (AD), known as DS-AD. How the increased APP gene dose acts and which APP products are responsible for DS-AD is not well understood, thus limiting strategies to target pathogenesis. As one approach to address this question, we used a novel class of γ-secretase modulators that promote γ-site cleavages by the γ-secretase complex, resulting in lower levels of the Aβ42 and Aβ40 peptides. METHODS Ts65Dn mice, which serve as a model of DS, were treated via oral gavage with 10 mg/kg/weekday of BPN15606 (a potent and novel pyridazine-containing γ-secretase modulators). Treatment started at 3 months-of-age and lasted for 4 months. RESULTS Demonstrating successful target engagement, treatment with BPN15606 significantly decreased levels of Aβ40 and Aβ42 in the cortex and hippocampus; it had no effect on full-length APP or its C-terminal fragments in either 2 N or Ts65Dn mice. Importantly, the levels of total amyloid-β were not impacted, pointing to BPN15606-mediated enhancement of processivity of γ-secretase. Additionally, BPN15606 rescued hyperactivation of Rab5, a protein responsible for regulating endosome function, and normalized neurotrophin signaling deficits. BPN15606 treatment also normalized the levels of synaptic proteins and tau phosphorylation, while reducing astrocytosis and microgliosis, and countering cognitive deficits. INTERPRETATION Our findings point to the involvement of increased levels of Aβ42 and/or Aβ40 in contributing to several molecular and cognitive traits associated with DS-AD. They speak to increased dosage of the APP gene acting through heightened levels of Aβ42 and/or Aβ40 as supporting pathogenesis. These findings further the interest in the potential use of γ-secretase modulators for treating and possibly preventing AD in individuals with DS. ANN NEUROL 2024;96:390-404.
Collapse
Affiliation(s)
- Xu-Qiao Chen
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Ann Becker
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Ricardo Albay
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Phuong D Nguyen
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Dmitry Karachentsev
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Amanda J Roberts
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, CA, USA
| | - Kevin D Rynearson
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - William C Mobley
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
28
|
El Brouzi MY, Lamtai M, Fath N, Rezqaoui A, Zghari O, El Hamzaoui A, Ibouzine-Dine L, El Hessni A, Mesfioui A. Exploring the neuroprotective role of melatonin against nickel-induced neurotoxicity in the left hippocampus. Biometals 2024:10.1007/s10534-024-00618-w. [PMID: 39060834 DOI: 10.1007/s10534-024-00618-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 06/15/2024] [Indexed: 07/28/2024]
Abstract
Previous studies have demonstrated that the hippocampus, a crucial region for memory and cognitive functions, is particularly vulnerable to adverse effects of exposure to heavy metals. Nickel (Ni) is a neurotoxic agent that, primarily induces oxidative stress, a process known to contribute to cellular damage, which consequently affects neurological functions. The antioxidant properties of melatonin are a promising option for preventing the adverse effects of Ni, especially by protecting cells against oxidative stress and related damage. In our investigation of the potential neuroprotective effects of melatonin against Ni-induced neurotoxicity, we chose to administer melatonin through intraperitoneal injection in rats following an intrahippocampal injection of Ni into the left hippocampus. This approach allows us a targeted investigation into the influence of melatonin on the neurotoxic effects of Ni, particularly within the crucial context of the hippocampus. In the present study, we demonstrated that melatonin efficiency reduced lactate dehydrogenase level, and preserved antioxidant enzyme activities in Ni-exposed hippocampal tissue. It also mitigated the decline in superoxide dismutase and catalase activities. On the other hand, melatonin could act directly by reducing reactive oxygen species Ni-induced overproduction. Taking to gather these two potential mechanisms of action could be responsible for the adverse effect of Ni on the behavioral alteration observed in our study. This study provides significant insights into the potential of melatonin to mitigate the detrimental effects of Ni on the brain, particularly into the hippocampal region, suggesting its possible implications for the treatment of neurological disorders related to Ni exposure.
Collapse
Affiliation(s)
- Mohamed Yassine El Brouzi
- Laboratory of Biology and Health, Neurosciences, Neuro-Immunology and Behaviour Unit, Faculty of Science, Ibn Tofail University, Kenitra, Morocco.
| | - Mouloud Lamtai
- Laboratory of Biology and Health, Neurosciences, Neuro-Immunology and Behaviour Unit, Faculty of Science, Ibn Tofail University, Kenitra, Morocco
| | - Nada Fath
- Compared Anatomy Unit, School of Veterinary Medicine, Hassan II Institute of Agronomy and Veterinary Medicine, Rabat, Morocco
- Physiology and Pathophysiology Laboratory, Department of Biology, Faculty of Sciences, Mohamed V University, Rabat, Morocco
| | - Ayoub Rezqaoui
- Laboratory of Biology and Health, Neurosciences, Neuro-Immunology and Behaviour Unit, Faculty of Science, Ibn Tofail University, Kenitra, Morocco
| | - Oussama Zghari
- Laboratory of Biology and Health, Neurosciences, Neuro-Immunology and Behaviour Unit, Faculty of Science, Ibn Tofail University, Kenitra, Morocco
| | - Abdelghafour El Hamzaoui
- Laboratory of Biology and Health, Neurosciences, Neuro-Immunology and Behaviour Unit, Faculty of Science, Ibn Tofail University, Kenitra, Morocco
| | - Laila Ibouzine-Dine
- Laboratory of Biology and Health, Neurosciences, Neuro-Immunology and Behaviour Unit, Faculty of Science, Ibn Tofail University, Kenitra, Morocco
| | - Aboubaker El Hessni
- Laboratory of Biology and Health, Neurosciences, Neuro-Immunology and Behaviour Unit, Faculty of Science, Ibn Tofail University, Kenitra, Morocco
| | - Abdelhalem Mesfioui
- Laboratory of Biology and Health, Neurosciences, Neuro-Immunology and Behaviour Unit, Faculty of Science, Ibn Tofail University, Kenitra, Morocco
| |
Collapse
|
29
|
Song HX, Xie YH, Fang YY, Lin JJ, Wang LL, Gan CL, Aschner M, Jiang YM. Sodium para-aminosalicylic acid attenuates combined manganese/iron-induced cortical synaptic damage in rats. Basic Clin Pharmacol Toxicol 2024; 135:81-97. [PMID: 38780039 DOI: 10.1111/bcpt.14033] [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: 12/24/2023] [Revised: 04/08/2024] [Accepted: 05/05/2024] [Indexed: 05/25/2024]
Abstract
We established experimental models of manganese (Mn) and iron (Fe) exposure in vitro and in vivo, and addressed the effects of manganese and iron combined exposure on the synaptic function of pheochromocytoma derived cell line 12 (PC12) cells and rat cortex, respectively. We investigated the protective effect of sodium para-aminosalicylate (PAS-Na) on manganese and iron combined neurotoxicity, providing a scientific basis for the prevention and treatment of ferromanganese combined neurotoxicity. Western blot and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) were performed to detect the expression levels of protein and mRNA related to synaptic damage. Y-maze novelty test and balance beam test were used to evaluate the motor and cognitive function of rats. Haematoxylin and eosin (H&E) and Nissl staining were performed to observe the cortical damage of rats. The results showed that the combined exposure of Mn and Fe in rats led to a synergistic effect, attenuating growth and development, and altering learning and memory as well as motor function. The combination of Mn and Fe also caused damage to the synaptic structure of PC12 cells, which is manifested as swelling of dendrites and axon terminals, and even lead to cell death. PAS-Na displayed some antagonistic effects against the Mn- and Fe-induced synaptic structural damage, growth, learning and memory impairment.
Collapse
Affiliation(s)
- Han-Xiao Song
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Yu-Han Xie
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Yuan-Yuan Fang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Jun-Jie Lin
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Lei-Lei Wang
- School of Public Health, Xiamen University, Xiamen, China
| | - Cui-Liu Gan
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Yue-Ming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| |
Collapse
|
30
|
Sun Y, Zhang H, Liu R, Huang R, Gao Z, Tian L, Zhu Y, Liu Y, Lu C, Wu L. Lancao decoction alleviates cognitive dysfunction: A new therapeutic drug and its therapeutic mechanism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155531. [PMID: 38492366 DOI: 10.1016/j.phymed.2024.155531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 02/22/2024] [Accepted: 03/10/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND Cognitive dysfunction (CD) is a neurodegenerative disease characterized primarily by the decline of learning and memory abilities. The physiological and pathological mechanisms of CD are very complex, which is mainly related to normal function of the hippocampus. Lancao decoction (LC) is a Chinese medicine formula, which has been used to treat neurodegenerative disorders. However, the potential of LC for the treatment of CD, as well as its underlying mechanisms, is unclear. PURPOSE In the study, we aimed to reveal the functional and neuronal mechanisms of LC's treatments for CD in scopolamine-induced mice. METHODS Gas chromatography (GC) was used to determine the stability of LC's extraction. CD model was established by the chronic induction of scopolamine (Scop, 1 mg/kg/day) for 1 week. Behavioral tests including morris water maze (MWM) and y-maze were used to evaluate learning and memory abilities of mice after LC's treatments. Immunofluorescence was used to detected the expressions of cFOS, Brdu and Ki67 after LC's treatments. Pharmacological blockade experiments explored the role of α-Amino-3‑hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) in LC's treatments for CD and its relationships with regeneration, activities and differentiation of neurons. RESULTS The results showed that LC was capable of improving spatial learning and memory and spontaneous alternating abilities in Scop-induced mice, which was similar to donepezil. LC could increase the number of cFOS positive cells, which was used as a marker of neuronal activity to upregulate by neuronal activities in hippocampus, but donepezil did not. Moreover, LC could strengthen neurogenesis and neuro-differentiation by increasing the number of Brdu and Ki67 positive cells in hippocampal dentate gyrus (DG), meanwhile, donepezil could only enhance the number of Ki67 positive cells. Transient inhibition of AMPAR by NBQX blunted the function of LC's treatment for CD and inhibited the enhanced effect of LC on Scop-induced hippocampal neuronal excitability and neurogenesis in mice. CONCLUSION To sum up, our study demonstrated that LC had the function of treating CD by enhancing content of acetylcholine (ACh) to activate AMPAR, which further up-regulated neurogenesis and neuronal differentiation to strengthen neuroactivities in hippocampus.
Collapse
Affiliation(s)
- Yan Sun
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Department of Pharmacy, Nanjing 210029, PR China; College of Chinese Medicine & College of Integrated Chinese and Western Medicine, Key Laboratory of Integrative Biomedicine for Brain Diseases, College of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Hailou Zhang
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders and School of Chinese Medicine, Jinan University, Guangzhou 510632, PR China.
| | - Ruiyi Liu
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders and School of Chinese Medicine, Jinan University, Guangzhou 510632, PR China
| | - Rumin Huang
- College of Chinese Medicine & College of Integrated Chinese and Western Medicine, Key Laboratory of Integrative Biomedicine for Brain Diseases, College of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Ziwei Gao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Department of Pharmacy, Nanjing 210029, PR China
| | - Liyuan Tian
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Department of Pharmacy, Nanjing 210029, PR China
| | - Yaping Zhu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Department of Pharmacy, Nanjing 210029, PR China
| | - Yuxin Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Department of Pharmacy, Nanjing 210029, PR China
| | - Chao Lu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Department of Pharmacy, Nanjing 210029, PR China
| | - Lei Wu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Department of Pharmacy, Nanjing 210029, PR China.
| |
Collapse
|
31
|
Fazekas CL, Török B, Correia P, Chaves T, Bellardie M, Sipos E, Horváth HR, Gaszner B, Dóra F, Dobolyi Á, Zelena D. The Role of Vesicular Glutamate Transporter Type 3 in Social Behavior, with a Focus on the Median Raphe Region. eNeuro 2024; 11:ENEURO.0332-23.2024. [PMID: 38839305 PMCID: PMC11154661 DOI: 10.1523/eneuro.0332-23.2024] [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: 08/30/2023] [Revised: 03/27/2024] [Accepted: 04/08/2024] [Indexed: 06/07/2024] Open
Abstract
Social behavior is important for our well-being, and its dysfunctions impact several pathological conditions. Although the involvement of glutamate is undeniable, the relevance of vesicular glutamate transporter type 3 (VGluT3), a specific vesicular transporter, in the control of social behavior is not sufficiently explored. Since midbrain median raphe region (MRR) is implicated in social behavior and the nucleus contains high amount of VGluT3+ neurons, we compared the behavior of male VGluT3 knock-out (KO) and VGluT3-Cre mice, the latter after chemogenetic MRR-VGluT3 manipulation. Appropriate control groups were included. Behavioral test battery was used for social behavior (sociability, social discrimination, social interaction, resident intruder test) and possible confounding factors (open field, elevated plus maze, Y-maze tests). Neuronal activation was studied by c-Fos immunohistochemistry. Human relevance was confirmed by VGluT3 gene expression in relevant human brainstem areas. VGluT3 KO mice exhibited increased anxiety, social interest, but also aggressive behavior in anxiogenic environment and impaired social memory. For KO animals, social interaction induced lower cell activation in the anterior cingulate, infralimbic cortex, and medial septum. In turn, excitation of MRR-VGluT3+ neurons was anxiolytic. Inhibition increased social interest 24 h later but decreased mobility and social behavior in aggressive context. Chemogenetic activation increased the number of c-Fos+ neurons only in the MRR. We confirmed the increased anxiety-like behavior and impaired memory of VGluT3 KO strain and revealed increased, but inadequate, social behavior. MRR-VGluT3 neurons regulated mobility and social and anxiety-like behavior in a context-dependent manner. The presence of VGluT3 mRNA on corresponding human brain areas suggests clinical relevance.
Collapse
Affiliation(s)
- Csilla Lea Fazekas
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, Pécs 7624, Hungary
- Institute of Experimental Medicine, Eötvös Loránd Research Network, Budapest 1084, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest 1085, Hungary
| | - Bibiána Török
- Institute of Experimental Medicine, Eötvös Loránd Research Network, Budapest 1084, Hungary
| | - Pedro Correia
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, Pécs 7624, Hungary
- Institute of Experimental Medicine, Eötvös Loránd Research Network, Budapest 1084, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest 1085, Hungary
| | - Tiago Chaves
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, Pécs 7624, Hungary
- Institute of Experimental Medicine, Eötvös Loránd Research Network, Budapest 1084, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest 1085, Hungary
| | - Manon Bellardie
- Institute of Experimental Medicine, Eötvös Loránd Research Network, Budapest 1084, Hungary
| | - Eszter Sipos
- Institute of Experimental Medicine, Eötvös Loránd Research Network, Budapest 1084, Hungary
| | - Hanga Réka Horváth
- Institute of Experimental Medicine, Eötvös Loránd Research Network, Budapest 1084, Hungary
| | - Balázs Gaszner
- Department of Anatomy, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, Pécs 7624, Hungary
| | - Fanni Dóra
- Human Brain Bank and Microdissection Laboratory, Semmelweis University, Budapest 1085, Hungary
- Laboratory of Neuromorphology, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest 1085, Hungary
| | - Árpád Dobolyi
- Laboratory of Neuromorphology, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest 1085, Hungary
- Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest 1117, Hungary
| | - Dóra Zelena
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, Pécs 7624, Hungary
- Institute of Experimental Medicine, Eötvös Loránd Research Network, Budapest 1084, Hungary
| |
Collapse
|
32
|
Kong H, Han YY, Yang GL, Li K, Yu L, Xie XK, Xia GY, Wei PJ, Zhang WR, Li CH. Tenuifolin improves learning and memory by regulating long-term potentiation and dendritic structure of hippocampal CA1 area in healthy female mice but not male mice. Behav Brain Res 2024; 466:114974. [PMID: 38554850 DOI: 10.1016/j.bbr.2024.114974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/02/2024]
Abstract
Polygala tenuifolia Wild is an ancient traditional Chinese medicine. Its main component, tenuifolin (TEN), has been proven to improve cognitive impairment caused by neurodegenerative diseases and ovariectomy. However, there was hardly any pharmacological research about TEN and its potential gender differences. Considering the reduction of TEN on learning and memory dysfunction in ovariectomized animals, therefore, we focused on the impact of TEN in different mice genders in the current study. Spontaneous alternation behavior (SAB), light-dark discrimination, and Morris water maze (MWM) tests were used to evaluate the mice's learning and memory abilities. The field excitatory postsynaptic potential (fEPSP) of the hippocampal CA1 region was recorded using an electrophysiological method, and the morphology of the dendritic structure was examined using Golgi staining. In the behavioral experiments, TEN improved the correct rate in female mice in the SAB test, the correct rate in the light-dark discrimination test, and the number of crossing platforms in the MWM test. Additionally, TEN reduced the latency of female mice rather than male mice in light-dark discrimination and MWM tests. Moreover, TEN could significantly increase the slope of fEPSP in hippocampal Schaffer-CA1 and enhance the total length and the number of intersections of dendrites in the hippocampal CA1 area in female mice but not in male mice. Collectively, the results of the current study showed that TEN improved learning and memory by regulating long-term potentiation (LTP) and dendritic structure of hippocampal CA1 area in female mice but not in males. These findings would help to explore the improvement mechanism of TEN on cognition and expand the knowledge of the potential therapeutic value of TEN in the treatment of cognitive impairment.
Collapse
Affiliation(s)
- Heng Kong
- School of Life Science, South China Normal University, Guangzhou, China
| | - Yuan-Yuan Han
- School of Life Science, South China Normal University, Guangzhou, China
| | - Gai-Ling Yang
- School of Life Science, South China Normal University, Guangzhou, China
| | - Kang Li
- School of Life Science, South China Normal University, Guangzhou, China
| | - Lu Yu
- School of Life Science, South China Normal University, Guangzhou, China
| | - Xun-Kai Xie
- School of Life Science, South China Normal University, Guangzhou, China
| | - Guang-Yuan Xia
- School of Life Science, South China Normal University, Guangzhou, China
| | - Peng-Ju Wei
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | | | - Chu-Hua Li
- School of Life Science, South China Normal University, Guangzhou, China.
| |
Collapse
|
33
|
Zong R, Zhang X, Dong X, Liu G, Zhang J, Gao Y, Zhang Z, Ma Y, Gao H, Gamper N. Genetic deletion of zinc transporter ZnT 3 induces progressive cognitive deficits in mice by impairing dendritic spine plasticity and glucose metabolism. Front Mol Neurosci 2024; 17:1375925. [PMID: 38807922 PMCID: PMC11130425 DOI: 10.3389/fnmol.2024.1375925] [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: 01/24/2024] [Accepted: 04/22/2024] [Indexed: 05/30/2024] Open
Abstract
Zinc transporter 3 (ZnT3) is abundantly expressed in the brain, residing in synaptic vesicles, where it plays important roles in controlling the luminal zinc levels. In this study, we found that ZnT3 knockout in mice decreased zinc levels in the hippocampus and cortex, and was associated with progressive cognitive impairments, assessed at 2, 6, and 9-month of age. The results of Golgi-Cox staining demonstrated that ZnT3 deficiency was associated with an increase in dendritic complexity and a decrease in the density of mature dendritic spines, indicating potential synaptic plasticity deficit. Since ZnT3 deficiency was previously linked to glucose metabolism abnormalities, we tested the expression levels of genes related to insulin signaling pathway in the hippocampus and cortex. We found that the Expression of glucose transporters, GLUT3, GLUT4, and the insulin receptor in the whole tissue and synaptosome fraction of the hippocampus of the ZnT3 knockout mice were significantly reduced, as compared to wild-type controls. Expression of AKT (A serine/threonine protein kinase) and insulin-induced AKT phosphorylation was also reduced in the hippocampus of ZnT3 knockout mice. We hypothesize that the ZnT3 deficiency and reduced brain zinc levels may cause cognitive impairment by negatively affecting glycose metabolism via decreased expression of key components of insulin signaling, as well as via changes in synaptic plasticity. These finding may provide new therapeutic target for treatments of neurodegenerative disorders.
Collapse
Affiliation(s)
- Rui Zong
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaoding Zhang
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaohui Dong
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Guan Liu
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jieyao Zhang
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yiting Gao
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhongyang Zhang
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yiming Ma
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Haixia Gao
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Nikita Gamper
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
34
|
McGill CJ, Christensen A, Qian W, Thorwald MA, Lugo JG, Namvari S, White OS, Finch CE, Benayoun BA, Pike CJ. Protection against APOE4 -associated aging phenotypes with the longevity-promoting intervention 17α-estradiol in male mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.12.584678. [PMID: 38559059 PMCID: PMC10980056 DOI: 10.1101/2024.03.12.584678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The apolipoprotein ε4 allele ( APOE4 ) is associated with decreased longevity, increased vulnerability to age-related declines, and disorders across multiple systems. Interventions that promote healthspan and lifespan represent a promising strategy to attenuate the development of APOE4 -associated aging phenotypes. Here we studied the ability of the longevity-promoting intervention 17α-estradiol (17αE2) to protect against age-related impairments in APOE4 versus the predominant APOE3 genotype using early middle-aged mice with knock-in of human APOE alleles. Beginning at age 10 months, male APOE3 or APOE4 mice were treated for 20 weeks with 17αE2 or vehicle then compared for indices of aging phenotypes body-wide. Across peripheral and neural measures, APOE4 was associated with poorer outcomes. Notably, 17αE2 treatment improved outcomes in a genotype-dependent manner favoring APOE4 mice. These data demonstrate a positive APOE4 bias in 17αE2-mediated healthspan actions, suggesting that longevity-promoting interventions may be useful in mitigating deleterious age-related risks associated with APOE4 genotype.
Collapse
|
35
|
Zhang T, Zhang Z, Geng J, Lin K, Lin X, Jiao M, Zhu J, Guo X, Lin Z. A New Approach for Exploring Reperfusion Brain Damage in Hypoxic Ischemic Encephalopathy. Mol Neurobiol 2024; 61:1417-1432. [PMID: 37721688 DOI: 10.1007/s12035-023-03645-9] [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: 02/13/2023] [Accepted: 09/05/2023] [Indexed: 09/19/2023]
Abstract
Reperfusion is an essential pathological stage in hypoxic ischemic encephalopathy (HIE). Although the Rice-Vannucci model is widely used in HIE research, it remains difficult to replicate HIE-related reperfusion brain injury. The purpose of this study is to establish a rat model of hypoxia ischemia reperfusion brain damage (HIRBD) using a common carotid artery (CCA) muscle bridge in order to investigate the mechanisms of cerebral resistance to hypoxic-ischemic and reperfusion brain damage. Random assignment of Sprague-Dawley (SD) rats to the Sham, HIRBD, and Rice-Vannucci groups. Changes in body weight, mortality rate, spontaneous alternation behavior test (SAB test), and dynamic changes in cerebral blood flow (CBF) were detected. The damaged cerebral cortices were extracted for morphological comparison, transcriptomic analysis, and quantitative real-time PCR. Harvesting the hippocampus for transmission electron microscopy (TEM) detection. As a result, CCA muscle bridge could effectively block CBF, which recovered after the muscle bridge detachment. Pathological comparison, the SAB test, and TEM analysis revealed that brain damage in Rice-Vannucci was more severe than HIRBD. Gpx1, S100a6, Cldn5, Esr1, and Gfap were highly expressed in both HIRBD and Rice-Vannucci. In conclusion, the CCA muscle bridge-established HIRBD model could be used as an innovative and dependable model to simulate pathological process of HIRBD.
Collapse
Affiliation(s)
- Tianlei Zhang
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zhiwei Zhang
- Second Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jiayi Geng
- Second Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Kexin Lin
- Second Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Xinru Lin
- Second Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Mengdie Jiao
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jianghu Zhu
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
- Key Laboratory of Children Genitourinary Diseases of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| | - Xiaoling Guo
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
- Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| | - Zhenlang Lin
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
- Second Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
- Key Laboratory of Children Genitourinary Diseases of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
36
|
Wan C, Xia Y, Yan J, Lin W, Yao L, Zhang M, Gaisler-Salomon I, Mei L, Yin DM, Chen Y. nNOS in Erbb4-positive neurons regulates GABAergic transmission in mouse hippocampus. Cell Death Dis 2024; 15:167. [PMID: 38396027 PMCID: PMC10891175 DOI: 10.1038/s41419-024-06557-1] [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: 07/12/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024]
Abstract
Neuronal nitric oxide synthase (nNOS, gene name Nos1) orchestrates the synthesis of nitric oxide (NO) within neurons, pivotal for diverse neural processes encompassing synaptic transmission, plasticity, neuronal excitability, learning, memory, and neurogenesis. Despite its significance, the precise regulation of nNOS activity across distinct neuronal types remains incompletely understood. Erb-b2 receptor tyrosine kinase 4 (ErbB4), selectively expressed in GABAergic interneurons and activated by its ligand neuregulin 1 (NRG1), modulates GABA release in the brain. Our investigation reveals the presence of nNOS in a subset of GABAergic interneurons expressing ErbB4. Notably, NRG1 activates nNOS via ErbB4 and its downstream phosphatidylinositol 3-kinase (PI3K), critical for NRG1-induced GABA release. Genetic removal of nNos from Erbb4-positive neurons impairs GABAergic transmission, partially rescued by the NO donor sodium nitroprusside (SNP). Intriguingly, the genetic deletion of nNos from Erbb4-positive neurons induces schizophrenia-relevant behavioral deficits, including hyperactivity, impaired sensorimotor gating, and deficient working memory and social interaction. These deficits are ameliorated by the atypical antipsychotic clozapine. This study underscores the role and regulation of nNOS within a specific subset of GABAergic interneurons, offering insights into the pathophysiological mechanisms of schizophrenia, given the association of Nrg1, Erbb4, Pi3k, and Nos1 genes with this mental disorder.
Collapse
Affiliation(s)
- Chaofan Wan
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Department of Rehabilitation, School of Health Science, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yucen Xia
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jinglan Yan
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Weipeng Lin
- Joint Center for Translational Medicine, Shanghai Fifth People's Hospital, Fudan University and School of Life Science, East China Normal University, Shanghai, 200062, China
| | - Lin Yao
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Meng Zhang
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Inna Gaisler-Salomon
- School of Psychological Sciences, The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa, 3498838, Israel
| | - Lin Mei
- Chinese Institute for Medical Research, Beijing, 100069, China
- Capital Medical University, Beijing, 100069, China
- Chinese Institute for Brain Research, Beijing, 102206, China
| | - Dong-Min Yin
- Joint Center for Translational Medicine, Shanghai Fifth People's Hospital, Fudan University and School of Life Science, East China Normal University, Shanghai, 200062, China.
| | - Yongjun Chen
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
- Guangdong Province Key Laboratory of Psychiatric Disorders, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
37
|
Onyekweli CC, Ben-Azu B, Oyovwi OM, Nwangwa EK, Ovuakporaye IS, Moke GE, Agbonifo-Chijiokwu E, Onome BO, Emojevwe V, Rotu AR. Epigallocatechin-gallate attenuates rapamycin exacerbated high fat diet-induced autophagy, hormonal dysregulation, testicular and brain oxidative stress, and neurochemical changes in rats. Food Chem Toxicol 2024; 184:114340. [PMID: 38097001 DOI: 10.1016/j.fct.2023.114340] [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: 09/30/2023] [Revised: 11/13/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023]
Abstract
This study investigated whether epigallocatechin-gallate (EGCG) could counteract the detrimental effects of high-fat diet (HFD)-induced obesity in rats exposed to rapamycin-induced reproductive and neuronal changes. Six rats per treatment group (n = 6) were utilized, in which groups 1 and 2 had dimethylsulfoxide (DMSO) (0.1%) and EGCG (80 mg/kg) respectively. Group 3 received HFD + 0.1% DMSO daily for 56 days. Group 4 received HFD + rapamycin (1 mg/kg) orally for 56 days. Rats in group 5 received HFD for 56 days and EGCG (80 mg/kg, p.o.) from days 29-56. Group 6 received the combination of HFD + rapamycin (56 days) with EGCG (80 mg/kg) from days 29-56. Cognitive loss was assessed using Y-maze-test (YMT). Afterwards, serum sex hormones, insulin-glucose balance, serotonin concentration, acetylcholinesterase activity, sperm features, antioxidants, and the markers of oxido-nitrergic, autophagy and apoptotic mediators were assessed. EGCG reversed rapamycin exacerbated HFD-induced alterations in spermatogenesis, insulin-glucose balance, reproductive hormones, oxido-nitrergic stress, and altered serotonin, acetylcholinesterase levels, and autophagic and apoptotic activities in rats' testes and brains respectively. EGCG significantly attenuated HFD-induced cognitive loss. The study showed that EGCG attenuated rapamycin-mediated HFD-induced spermatogenesis deficiency and cognitive impairment via normalization of reproductive hormones, testicular and brain oxidative stress, apoptotic, autophagic activities, with serotonin and cholinergic levels in rats.
Collapse
Affiliation(s)
- Chinedu Charles Onyekweli
- Department of Physiology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| | - Benneth Ben-Azu
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria.
| | - O Mega Oyovwi
- Department of Physiology, Faculty of Basic Medical Sciences, Adeleke University, Ede, Osun State, Nigeria
| | - E Kingsley Nwangwa
- Department of Physiology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria.
| | - I Simon Ovuakporaye
- Department of Physiology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| | - Goodies Emuesiri Moke
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| | - Ejime Agbonifo-Chijiokwu
- Department of Physiology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| | - B Oghenetega Onome
- Department of Physiology, School of Basic Medical Sciences, Babcock University, Ilisan-Remo, Ogun State, Nigeria
| | - Victor Emojevwe
- Department of Physiology, University of Medical Sciences, Ondo State, Nigeria
| | - A Rume Rotu
- Department of Physiology, University, Ibadan, Oyo State, Nigeria
| |
Collapse
|
38
|
Gannon O, Tremble SM, McGinn C, Guth R, Scoppettone N, Hunt RD, Prakash K, Johnson AC. Angiotensin II-mediated hippocampal hypoperfusion and vascular dysfunction contribute to vascular cognitive impairment in aged hypertensive rats. Alzheimers Dement 2024; 20:890-903. [PMID: 37817376 PMCID: PMC10917018 DOI: 10.1002/alz.13491] [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: 07/24/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 10/12/2023]
Abstract
INTRODUCTION Chronic hypertension increases the risk of vascular cognitive impairment (VCI) by ∼60%; however, how hypertension affects the vasculature of the hippocampus remains unclear but could contribute to VCI. METHODS Memory, hippocampal perfusion, and hippocampal arteriole (HA) function were investigated in male Wistar rats or spontaneously hypertensive rats (SHR) in early (4 to 5 months old), mid (8 to 9 months old), or late adulthood (14 to 15 months old). SHR in late adulthood were chronically treated with captopril (angiotensin converting enzyme inhibitor) or apocynin (antioxidant) to investigate the mechanisms by which hypertension contributes to VCI. RESULTS Impaired memory in SHR in late adulthood was associated with HA endothelial dysfunction, hyperconstriction, and ∼50% reduction in hippocampal blood flow. Captopril, but not apocynin, improved HA function, restored perfusion, and rescued memory function in aged SHR. DISCUSSION Hippocampal vascular dysfunction contributes to hypertension-induced memory decline through angiotensin II signaling, highlighting the therapeutic potential of HAs in protecting neurocognitive health later in life. HIGHLIGHTS Vascular dysfunction in the hippocampus contributes to vascular cognitive impairment. Memory declines with age during chronic hypertension. Angiotensin II causes endothelial dysfunction in the hippocampus in hypertension. Angiotensin II-mediated hippocampal arteriole dysfunction reduces blood flow. Vascular dysfunction in the hippocampus impairs perfusion and memory function.
Collapse
Affiliation(s)
- Olivia Gannon
- Department of Neurological SciencesUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| | - Sarah M. Tremble
- Department of Neurological SciencesUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| | - Conor McGinn
- Department of Neurological SciencesUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| | - Ruby Guth
- Department of Neurological SciencesUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| | - Nadia Scoppettone
- Department of Neurological SciencesUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| | - Ryan D. Hunt
- Department of Neurological SciencesUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| | - Kirtika Prakash
- Department of Neurological SciencesUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| | - Abbie C. Johnson
- Department of Neurological SciencesUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| |
Collapse
|
39
|
Yi Y, Zhang Y, Song Y, Lu Y. Treadmill Running Regulates Adult Neurogenesis, Spatial and Non-spatial Learning, Parvalbumin Neuron Activity by ErbB4 Signaling. Cell Mol Neurobiol 2024; 44:17. [PMID: 38285192 PMCID: PMC11407172 DOI: 10.1007/s10571-023-01439-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 11/06/2023] [Indexed: 01/30/2024]
Abstract
Exercise can promote adult neurogenesis and improve symptoms associated with schizophrenia and other mental disorders via parvalbumin (PV)-positive GABAergic interneurons in the dentate gyrus ErbB4 is the receptor of neurotrophic factor neuregulin 1, expressed mostly in PV-positive interneurons. Whether ErbB4 in PV-positive neurons mediates the beneficial effect of exercise and adult neurogenesis on mental disorder needs to be further investigation. Here, we first conducted a four-week study on the effects of AG1478, an ErbB4 inhibitor, on memory and neurogenesis. AG1478 significantly impaired the performance in several memory tasks, including the T-maze, Morris water maze, and contextual fear conditioning, downregulated the expression of total ErbB4 (T-ErbB4) and the ratio of phosphate-ErbB4 (p-ErbB4) to T-ErbB4, and associated with neurogenesis impairment. Interestingly, AG1478 also appeared to decrease intracellular calcium levels in PV neurons, which could be reversed by exercise. These results suggest exercise may regulate adult neurogenesis and PV neuron activity through ErbB4 signaling. Overall, these findings provide further evidence of the importance of exercise for neurogenesis and suggest that targeting ErbB4 may be a promising strategy for improving memory and other cognitive functions in individuals with mental disorders.
Collapse
Affiliation(s)
- Yandong Yi
- Department of Pharmacy, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Physiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuejin Zhang
- Department of Physiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, 430030, China
- Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuanlong Song
- Department of Physiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, 430030, China
- Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yisheng Lu
- Department of Physiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
40
|
Zhu B, Cao A, Chen C, Zhou W, Luo W, Gui Y, Wang Q, Xu Z, Wang J. MMP-9 inhibition alleviates postoperative cognitive dysfunction by improving glymphatic function via regulating AQP4 polarity. Int Immunopharmacol 2024; 126:111215. [PMID: 38000234 DOI: 10.1016/j.intimp.2023.111215] [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: 09/15/2023] [Revised: 10/31/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023]
Abstract
Postoperative cognitive dysfunction (POCD) is a common complication after surgery, characterized by deficits in memory, attention and cognitive flexibility. However, the underlying mechanisms of POCD remain unclear. Neuroinflammation and blood-brain barrier disruption have been implicated as potential pathological processes. This study explores the neuroprotective effects and mechanisms of the matrix metalloproteinase(MMP-9)inhibitor GM6001 against POCD. We hypothesize GM6001 may reduce neuroinflammation and preserve blood-brain barrier integrity through direct inhibition of MMP-9. Moreover, GM6001 may stabilize aquaporin-4 polarity and glymphatic clearance function by modulating MMP-9-mediated cleavage of dystroglycan, a key protein for aquaporin-4 anchoring. Our results demonstrate GM6001 alleviates postoperative cognitive deficits and neuroinflammation. GM6001 also preserves blood-brain barrier integrity and rescues aquaporin-4 mislocalization after surgery. This study reveals a novel dual role for MMP-9 inhibition in cognitive protection through direct anti-neuroinflammatory effects and regulating aquaporin-4 membrane distribution. Targeting MMP-9 may represent a promising strategy to prevent postoperative cognitive dysfunction by integrating multiple protective mechanisms.
Collapse
Affiliation(s)
- Binbin Zhu
- The First Affiliated Hospital of Ningbo University, Ningbo 315000, China; Health Science Center, Ningbo University, Ningbo 315000, China
| | - Angyang Cao
- The First Affiliated Hospital of Ningbo University, Ningbo 315000, China; Health Science Center, Ningbo University, Ningbo 315000, China
| | - Chunqu Chen
- The First Affiliated Hospital of Ningbo University, Ningbo 315000, China; Health Science Center, Ningbo University, Ningbo 315000, China
| | - Weijian Zhou
- The First Affiliated Hospital of Ningbo University, Ningbo 315000, China; Health Science Center, Ningbo University, Ningbo 315000, China
| | - Wenjun Luo
- The First Affiliated Hospital of Ningbo University, Ningbo 315000, China; Health Science Center, Ningbo University, Ningbo 315000, China
| | - Yu Gui
- The First Affiliated Hospital of Ningbo University, Ningbo 315000, China
| | - Qinwen Wang
- Health Science Center, Ningbo University, Ningbo 315000, China
| | - Zhipeng Xu
- The First Affiliated Hospital of Ningbo University, Ningbo 315000, China
| | - Jianhua Wang
- The First Affiliated Hospital of Ningbo University, Ningbo 315000, China; Health Science Center, Ningbo University, Ningbo 315000, China.
| |
Collapse
|
41
|
Liu H, Gong Z, Li Z, Ye T, Cao A, He S, Lin S, Duan J, Lin X. Distribution, connection and function of ALDH1A1 +/TH + neurons in substantia nigra pars reticulata of mouse. Neurosci Lett 2024; 818:137555. [PMID: 37972684 DOI: 10.1016/j.neulet.2023.137555] [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: 09/18/2023] [Revised: 10/25/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023]
Abstract
The massive cell death of dopaminergic neurons (DANs) in substantia nigra pars compacta (SNC) is associated with motor diseases, such as Parkinson's disease. Moreover, as a subtype of DANs in SNC, ALDH1A1+ neurons show better resistance to PD related neurotoxin. DANs can also be found in the substantia nigra pars reticulata (SNR), however, whether they are ALDH1A1+ neurons are rarely reported, as well as their projection, function, and reaction in the PD pathology. We studied the distribution of ALDH1A1+ neurons and track their projection by injecting pAAV. We figured out that, in SNR, 87 % neurons are ALDH1A1+/TH+ in ALDH1A1+ cluster averagely, while ALDH1A1+/TH+: TH+ is 52 % averagely. There are two enrichment regions of ALDH1A1+/TH+ neurons at brgma -3.40 mm and brgma -3.70 mm in the SNR of the nTg mice. Nevertheless, in one type of PD-liked mice model, the proportion of ALDH1A1+/TH+: ALDH1A1+ neurons are 98 % averagely, while ALHD1A1+/TH+: TH+ is 57 %. Intriguingly, neuro-tracing discovered that there may be a previously unreported connection between SNR and anterior dorsal thalamus (ADT). The mouse received MPTP stereotactic injection to destroy TH+ neurons in SNR showed depression behavior, indicated the DANs death in SNR may contribute to depression behavior.
Collapse
Affiliation(s)
- Hao Liu
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China; Department of Human Anatomy and physiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China
| | - Zhuo Gong
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China; Department of Human Anatomy and physiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China
| | - Zhao Li
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China; Department of Human Anatomy and physiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China
| | - Tonglin Ye
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China; Department of Human Anatomy and physiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China
| | - Anqi Cao
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China; Department of Human Anatomy and physiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China
| | - Shuaiying He
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China; Department of Human Anatomy and physiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China
| | - Sijia Lin
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China; Department of Human Anatomy and physiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China
| | - Jinhai Duan
- Eastern Department of Neurology, Guangdong Geriatrics Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Xian Lin
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China; Department of Human Anatomy and physiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China.
| |
Collapse
|
42
|
Park H, Kwon HS, Lee KY, Kim YE, Son JW, Choi NY, Lee EJ, Han MH, Park DW, Kim S, Koh SH. GV1001 modulates neuroinflammation and improves memory and behavior through the activation of gonadotropin-releasing hormone receptors in a triple transgenic Alzheimer's disease mouse model. Brain Behav Immun 2024; 115:295-307. [PMID: 37884161 DOI: 10.1016/j.bbi.2023.10.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 09/22/2023] [Accepted: 10/22/2023] [Indexed: 10/28/2023] Open
Abstract
GV1001 protects neural cells from amyloid-β (Aβ) toxicity and other stressors in in vitro studies and demonstrates clinically beneficial effects in patients with moderate to severe Alzheimer's disease (AD). Here, we investigated the protective effects and mechanism of action of GV1001 in triple transgenic AD (3xTg-AD) mice. We found that GV1001 improved memory and cognition in middle- and old-aged 3xTg-AD mice. Additionally, it reduced Aβ oligomer and phospho-tau (Ser202 and Thr205) levels in the brain, and mitigated neuroinflammation by promoting a neuroprotective microglial and astrocyte phenotype while diminishing the neurotoxic ones. In vitro, GV1001 bound to gonadotropin releasing hormone receptors (GnRHRs) with high affinity. Levels of cyclic adenosine monophosphate, a direct downstream effector of activated GnRHRs, increased after GV1001 treatment. Furthermore, inhibition of GnRHRs blocked GV1001-induced effects. Thus, GV1001 might improve cognitive and memory functions of 3xTg-AD mice by suppressing neuroinflammation and reducing Aβ oligomers levels and phospho-tau by activating GnRHRs and their downstream signaling pathways.
Collapse
Affiliation(s)
- Hyunhee Park
- Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Hyuk Sung Kwon
- Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Kyu-Yong Lee
- Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Ye Eun Kim
- Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Jeong-Woo Son
- Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Na-Young Choi
- Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Eun Ji Lee
- Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Myung-Hoon Han
- Department of Neurosurgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Dong Woo Park
- Department of Radiology, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Sangjae Kim
- Teloid Inc., 3580 Wilshire Boulevard, Suite 900-31, Los Angeles, CA 90010, USA.
| | - Seong-Ho Koh
- Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea; Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul 04763, South Korea.
| |
Collapse
|
43
|
Mahnashi MH, Ayaz M, Alqahtani YS, Alyami BA, Shahid M, Alqahtani O, Kabrah SM, Zeb A, Ullah F, Sadiq A. Quantitative-HPLC-DAD polyphenols analysis, anxiolytic and cognition enhancing potentials of Sorbaria tomentosa Lindl. Rehder. JOURNAL OF ETHNOPHARMACOLOGY 2023; 317:116786. [PMID: 37328081 DOI: 10.1016/j.jep.2023.116786] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/01/2023] [Accepted: 06/12/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Medicinal plants of the family Rosaceae have a long history of traditional uses in the management of neurological disorders. Sorbaria tomentosa Lindl. Rehder is composed of antioxidant and neuroprotective polyphenolics. AIMS OF THE STUDY The current study was designed to explore phenolics profile via high performance liquid chromatography-photodiode array detector (HPLC-DAD) and validated the neuroprotective and anxiolytic potentials of S. tomentosa by applying in vitro and in vivo approaches. MATERIALS AND METHODS The plant crude methanolic extract (St.Crm) and fractions were subjected to HPLC-DAD analysis for qualitative and quantitative assessment of phytochemicals. Samples were screened for in vitro free radicals scavenging assays by using 2,2-diphenylpicrylhydrazyl (DPPH), 2,2'-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) along with acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) enzymes inhibition assays. For cognitive and anxiolytic studies, mice were subjected to open field, elevated plus maze (EPM), light-dark model, Y-maze, shallow water maze (SWM), and novel object recognition (NOR) tests. RESULTS HPLC-DAD analysis revealed the presence of high concentrations of phenolic compounds. For instance, in St.Cr, 21 phenolics were quantified, among which apigenin-7-glucoside (291.6 mg/g), quercetin (122.1 mg/g), quercetin-3-feruloylsophoroside-7-glucoside (52.6 mg/g), quercetin-7-glucoside (51.8 mg/g), ellagic acid (42.7 mg/g), luteolin (45.0 mg/g), kaempferol (40.5 mg/g), 5-feruloylquinic acid (43.7 mg/g) were present in higher concentrations. Likewise, in ethyl acetate fraction (St.Et.Ac), 21 phenolics were identified as 3,5-di-caffeoylquinic acid (177.4 mg/g) and 5-hydroxybenzoylquinic acid (46.9 mg/g) were most abundant phytochemicals. Highly valuable phenolics were also identified in other fractions including butanol (St.Bt), chloroform (St.Chf), and n-hexane (St.Hex). The various fractions exhibited concentration dependent inhibition of free radicals in DPPH and ABTS assays. Potent AChE inhibitory potentials were revealed by the test samples with St.Chf, St.Bt and St.EtAc being the most active having an IC50 of 298.1, 580.1, and 606.47 μg mL-1, respectively. Similarly, St.Chf, St.Bt, St.EtAc and St.Cr exhibited potent BChE inhibitory activity and was observed as 59.14, 54.73, 51.35 and 49.44%, respectively. A significant improvement in the exploratory behavior was observed in open field test and stress/anxiety was relieved effectively at 50-100 mg/kg. Likewise, EPM, light-dark and NOR tests revealed an anxiolytic and memory enhancing behaviors. These effects were further corroborated from the Y-maze and SWM transgenic studies that showed considerable improvement in cognition retention. CONCLUSIONS These findings concluded that S. tomentosa possessed potential anxiolytic and nootropic efficacies and may have therapeutic potential in neurodegenerative disorders.
Collapse
Affiliation(s)
- Mater H Mahnashi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia.
| | - Muhammad Ayaz
- Department of Pharmacy, Faculty of Biological Sciences, University of Malakand, Chakdara, 18000, Dir (L), KP, Pakistan.
| | - Yahya S Alqahtani
- Department of Pharmaceutical Chemistry, College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia.
| | - Bandar A Alyami
- Department of Pharmaceutical Chemistry, College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia.
| | - Muhammad Shahid
- Department of Pharmacy, CECOS University of IT and Emerging Sciences, Peshawar, Pakistan.
| | - Omaish Alqahtani
- Department of Pharmacognosy, College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia.
| | - Saeed M Kabrah
- Department of Laboratory Medicine Faculty of Applied Medical Sciences, Umm Al-Qura University, Kingdom of Saudi Arabia.
| | - Alam Zeb
- Department of Biochemistry, University of Malakand, Chakdara, 18000, Dir (L), KP, Pakistan.
| | - Farhat Ullah
- Department of Pharmacy, Faculty of Biological Sciences, University of Malakand, Chakdara, 18000, Dir (L), KP, Pakistan.
| | - Abdul Sadiq
- Department of Pharmacy, Faculty of Biological Sciences, University of Malakand, Chakdara, 18000, Dir (L), KP, Pakistan.
| |
Collapse
|
44
|
Alam A, Ali G, Nawaz A, AlOmar TS, Rauf A, Ayaz M, Ahmad S, Almasoud N, AlOmar AS, Khalil AA, Wilairatana P. Neuroprotective evaluation of diospyrin against drug-induced Alzheimer's disease. Fitoterapia 2023; 171:105703. [PMID: 37852388 DOI: 10.1016/j.fitote.2023.105703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 10/20/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease linked to memory impairment. A current investigation was performed to assess the neuroprotective effect of Diospyrin, a novel therapeutic agent, for the curing of Alzheimer's disease. For this purpose, in-vitro acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) inhibitory assays and antioxidant studies were conducted, whereas in-vivo studies involved different behavioral animal models tests such as elevated plus maze (EPM), morris water maze (MWM) and paddling Y-maze test. Results of the in-vitro analysis showed IC50 values of 95 μg/mL for AChE and 110 μg/mL for BChE as compared to the standard drug donepezil (IC50: 95 & 85 μg/mL, respectively). DPPH antioxidant assay showed a maximum of 72.85% inhibition (IC50: 139.74 μg/mL) of DPPH-free radicals at the highest concentration of 1000 μg/mL as compared to the ascorbic acid (IC50: 13.72 μg/mL). Moreover, the in-vivo analysis revealed that diospyrin treatment demonstrated gradual betterment in memory and enhanced motor functionality. On the other hand, the computational analysis also showed that the diospyrin had exceptional binding affinities for both AChE and BChE enzymes. In the net shell, it may be deduced that our compound diospyrin could be a valuable drug candidate in managing neurodegenerative disorders like AD.
Collapse
Affiliation(s)
- Aftab Alam
- Department of Pharmacy, University of Peshawar, Peshawar 25120, Pakistan
| | - Gowhar Ali
- Department of Pharmacy, University of Peshawar, Peshawar 25120, Pakistan.
| | - Asif Nawaz
- Department of Pharmacy, Faculty of Biological Sciences, University of Malakand, Chakdara 18000, Pakistan
| | - Taghrid S AlOmar
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84427, Riyadh 11671, Saudi Arabia
| | - Abdur Rauf
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84427, Riyadh 11671, Saudi Arabia; Department of Chemistry, University of Swabi, Anbar-23561, Khyber, Pakhtunkhwa, Pakistan.
| | - Muhammad Ayaz
- Department of Pharmacy, Faculty of Biological Sciences, University of Malakand, Chakdara 18000, Pakistan
| | - Sajjad Ahmad
- Department of Health and Biological Sciences, Abasyn University, Peshawar, Pakistan
| | - Najla Almasoud
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84427, Riyadh 11671, Saudi Arabia
| | - Abdulaziz S AlOmar
- Department of Family Medicine, College of Medicine, Al-Imam Mohammad ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Anees Ahmed Khalil
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahore, Pakistan
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
45
|
Wang H, Bullert AJ, Li X, Stevens H, Klingelhutz AJ, Ankrum JA, Adamcakova-Dodd A, Thorne PS, Lehmler HJ. Use of a polymeric implant system to assess the neurotoxicity of subacute exposure to 2,2',5,5'-tetrachlorobiphenyl-4-ol, a human metabolite of PCB 52, in male adolescent rats. Toxicology 2023; 500:153677. [PMID: 37995827 PMCID: PMC10757425 DOI: 10.1016/j.tox.2023.153677] [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: 09/05/2023] [Revised: 11/06/2023] [Accepted: 11/18/2023] [Indexed: 11/25/2023]
Abstract
Polychlorinated biphenyls (PCBs) are persistent organic pollutants (POPs) that ubiquitously exist in the environment. PCB exposure has been linked to cancer and multi-system toxicity, including endocrine disruption, immune inhibition, and reproductive and neurotoxicity. 2,2',5,5'-Tetrachlorobiphenyl (PCB 52) is one of the most frequently detected congeners in the environment and human blood. The hydroxylated metabolites of PCB 52 may also be neurotoxic, especially for children whose brains are still developing. However, it is challenging to discern the contribution of these metabolites to PCB neurotoxicity because the metabolism of PCB is species-dependent. In this study, we evaluated the subacute neurotoxicity of a human-relevant metabolite, 2,2',5,5'-tetrachlorobiphenyl-4-ol (4-52), on male adolescent Sprague Dawley rats, via a novel polymeric implant drug delivery system grafted subcutaneously, at total loading concentrations ranging from 0%, 1%, 5%, and 10% of the implant (w/w) for 28 days. Y-maze, hole board test, open field test, and elevated plus maze were performed on exposure days 24-28 to assess their locomotor activity, and exploratory and anxiety-like behavior. 4-52 and other possible hydroxylated metabolites in serum and vital tissues were quantified using gas chromatography with tandem mass spectrometry (GC-MS/MS). Our results demonstrate the sustained release of 4-52 from the polymeric implants into the systemic circulation in serum and tissues. Dihydroxylated and dechlorinated metabolites were detected in serum and tissues, depending on the dose and tissue type. No statistically significant changes were observed in the neurobehavioral tasks across all exposure groups. The results demonstrate that subcutaneous polymeric implants provide a straightforward method to expose rats to phenolic PCB metabolites to study neurotoxic outcomes, e.g., in memory, anxiety, and exploratory behaviors.
Collapse
Affiliation(s)
- Hui Wang
- Department of Occupational and Environmental Health, the University of Iowa, Iowa City, IA, USA
| | - Amanda J Bullert
- Department of Occupational and Environmental Health, the University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Neuroscience, the University of Iowa, Iowa City, IA, USA
| | - Xueshu Li
- Department of Occupational and Environmental Health, the University of Iowa, Iowa City, IA, USA
| | - Hanna Stevens
- Interdisciplinary Graduate Program in Neuroscience, the University of Iowa, Iowa City, IA, USA; Department of Psychiatry, the University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Human Toxicology, the University of Iowa, Iowa City, IA, USA
| | | | - James A Ankrum
- Roy J. Carver Department of Biomedical Engineering, the University of Iowa, Iowa City, IA, USA
| | - Andrea Adamcakova-Dodd
- Department of Occupational and Environmental Health, the University of Iowa, Iowa City, IA, USA
| | - Peter S Thorne
- Department of Occupational and Environmental Health, the University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Neuroscience, the University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Human Toxicology, the University of Iowa, Iowa City, IA, USA
| | - Hans-Joachim Lehmler
- Department of Occupational and Environmental Health, the University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Neuroscience, the University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Human Toxicology, the University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
46
|
Salama A, Elgohary R. Influence of chrysin on D-galactose induced-aging in mice: Up regulation of AMP kinase/liver kinase B1/peroxisome proliferator-activated receptor-γ coactivator 1-α signaling pathway. Fundam Clin Pharmacol 2023; 37:947-959. [PMID: 36977287 DOI: 10.1111/fcp.12895] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 02/24/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023]
Abstract
Adenosine monophosphate kinase/liver kinase B1/peroxisome proliferator-activated receptor-γ coactivator 1-α (AMPK/LKB1/PGC1α) pathway has a vital role in regulating age-related diseases. It controls neurogenesis, cell proliferation, axon outgrowth, and cellular energy homeostasis. AMPK pathway also regulates mitochondrial synthesis. The current study evaluated the effect of chrysin on D-galactose (D-gal) induced-aging, neuron degeneration, mitochondrial dysfunction, oxidative stress, and neuroinflammation in mice. The mice were allocated randomly into four groups (10 each group): Group 1: normal control group, Group 2: D-gal group, Groups 3 and 4: chrysin (125 and 250 mg/kg, respectively). Groups 2-4 were injected with D-gal (200 mg/kg/day; s.c) for 8 weeks to induce aging. Groups 3 and 4 were orally gavaged every day concurrent with D-gal. At the end of experiment, behavioral, brain biochemical and histopathological changes were monitored. Chrysin administration elevated discrimination ratio in object recognition, Y Maze percentage alternation, locomotor activity and brain contents of AMPK, LKB1, PGC1α, NAD (P)H quinone oxidoreductase 1 (NQO1), heme oxygenase 1 (HO-1), nerve growth factor (NGF) (neurotrophin-3; NT-3), and seretonin as well as reduced brain contents of tumor necrosis factor-alpha (TNF-α), nuclear factor kappa B (NF-κB), advanced glycation end products (AGEs) and glial fibrillary acidic protein (GFAP) compared to D-gal-treated mice. Chrysin also alleviated cerebral cortex and white matter neurons degeneration. Chrysin protects against neurodegeneration, improves mitochondrial autophagy and biogenesis as well as activates antioxidant genes expression. In addition, chrysin ameliorates neuroinflammation and stimulates the release of NGF and serotonin neurotransmitter. So, chrysin has a neuroprotective effect in D-gal induced-aging in mice.
Collapse
Affiliation(s)
- Abeer Salama
- Pharmacology Department, National Research Centre, El-Buhouth St., Cairo, Dokki, 12622, Egypt
| | - Rania Elgohary
- Narcotics, Ergogenics and Poisons Department, National Research Centre, El-Buhouth St., Cairo, Dokki, 12622, Egypt
| |
Collapse
|
47
|
Rezqaoui A, Ibouzine-Dine L, Elhamzaoui A, Brouzi MYE, Dimaoui A, Hessni AE, Mesfioui A. Potential Role of Oxidative Stress in the Effects of Chronic Administration of Iron on Affective and Cognitive Behavior on Male Wistar Rat. Biol Trace Elem Res 2023; 201:4812-4826. [PMID: 36683122 DOI: 10.1007/s12011-023-03560-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/05/2023] [Indexed: 01/24/2023]
Abstract
In this work, we studied the impact of chronic iron exposure, in the form of iron sulfate (FeSo4), on affective and cognitive disorders and oxidative stress in the male Wistar rat. The treatment was carried out for 8 weeks, the rats received an intraperitoneal injection of iron at different doses: 0.25, 0.5, and 1 mg/kg. Affective and cognitive disorders are assessed in open field test (OFT), elevated plus maze (EPM), forced swimming test (FST), Morris water maze (MWM), and Y-maze. The hippocampus and prefrontal cortex of each animal were taken for biochemical examination. Our results show that iron exerts anxiogenic and depressogenic effects, which were observed first at the dose of 0.5 mg/kg and continued in a dose-dependent manner up to the maximum tested dose of 1 mg/kg. According to results from the MWM and Y-maze tests, continuous exposure to iron induces cognitive disorders that are defined by the disturbance of working memory and influences spatial learning performance causing a deficit of spatial memory retention. We noted that chronic exposure to iron can be associated with the appearance of a state of oxidative stress in the hippocampus and the prefrontal cortex demonstrated by an increase in lipid peroxidation, an increase in nitric oxide, and also by disturbances in the antioxidant defense systems following a determination of the concentrations of catalase. In conclusion, we can deduce from this work that chronic iron exposure can be related to the induction of cognitive and affective disorders and oxidative stress.
Collapse
Affiliation(s)
- Ayoub Rezqaoui
- Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco.
| | - Laila Ibouzine-Dine
- Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco
| | - Abdelghafour Elhamzaoui
- Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco
| | - Mohamed Yassine El Brouzi
- Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco
| | - Amal Dimaoui
- Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco
| | - Aboubaker El Hessni
- Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco
| | - Abdelhalem Mesfioui
- Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco
| |
Collapse
|
48
|
Kim J, Kang H, Lee YB, Lee B, Lee D. A quantitative analysis of spontaneous alternation behaviors on a Y-maze reveals adverse effects of acute social isolation on spatial working memory. Sci Rep 2023; 13:14722. [PMID: 37679447 PMCID: PMC10485067 DOI: 10.1038/s41598-023-41996-4] [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: 05/08/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023] Open
Abstract
Animals tend to alternate between different choices, which requires the ability to remember recent choices. The Y-maze spontaneous alternation test is widely used in various animal models for assessing short-term memory, and its precise evaluation depends upon the accurate determination of the arm visit sequence. However, an objective method for defining arm visits is lacking owing to uncertainty regarding the extent to which an animal must go into the arm to be considered visited. Here, we conducted quantitative analyses on mice behavior in the Y-maze while systematically varying the arm visit threshold and assessed the effect of acute social isolation on spatial working memory. Our results revealed that 24-h social isolation significantly reduced spontaneous alternation rate when the arm threshold was set at the distal part of the arm. Furthermore, the memory of the recently visited arms faded away faster in the socially isolated mice. However, other behavioral factors were comparable to those of the group-housed mice, indicating a specific impairment of short-term memory. Our findings suggest that the location of arm visit threshold is critical for the precise evaluation of short-term memory, and our study provides a method for comprehensively and systematically assessing spontaneous alternation behavior in the Y-maze.
Collapse
Affiliation(s)
- Joowon Kim
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Korea
| | - Hyeyeon Kang
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Korea
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Korea
| | - Young-Beom Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Korea
| | - Boyoung Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Korea
| | - Doyun Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Korea.
| |
Collapse
|
49
|
Salemi S, Zamanian MY, Giménez‐Llort L, Jalali Z, Mahmoodi M, Golmohammadi M, Kaeidi A, Taghipour Z, Khademalhosseini M, Modanloo M, Hajizadehi MR. Distinct signatures on d-galactose-induced aging and preventive/protective potency of two low-dose vitamin D supplementation regimens on working memory, muscular damage, cardiac and cerebral oxidative stress, and SIRT1 and calstabin2 downregulation. Food Sci Nutr 2023; 11:5050-5062. [PMID: 37701236 PMCID: PMC10494626 DOI: 10.1002/fsn3.3422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/21/2023] [Accepted: 04/22/2023] [Indexed: 09/14/2023] Open
Abstract
Chronic administration of d-galactose (d-gal) in rodents reproduces the overproduction of reactive oxygen species of physiological aging. The present research shows for the first time distinct signatures on d-gal-induced aging (500 mg/kg, 6 weeks) and the preventive and protective potential of two vitamin D (50 IU) supplementation regimens (pre-induction and simultaneous, respectively) in two vital organs (heart and brain). d-gal-induced notorious alterations in working memory, a strong increase in brain malondialdehyde (MDA) oxidative levels, and strong downregulation of sirtuin 1 (SIRT1) in the heart and hippocampus and of calstabin2 in the heart. Cardiac and brain superoxide dismutase (SOD) and glutathione peroxidase (GPx) enzymatic antioxidant capacities were damaged, brain calstabin2 was downregulated, and neuropathology was observed. Heart damage also included a moderate increase in MDA levels, serologic lactate dehydrogenase (LDH), total creatine kinase (CK) activities, and histopathological alterations. The used dose of vitamin D was enough to prevent cognitive impairment, avoid muscular damage, hamper cardiac and cerebral oxidative stress, and SIRT1 and calstabin2 downregulation. Most importantly, the potencies of the two preventive schedules depended on the tissue and level of study. The pre-induction schedule prevented d-gal-induced aging by 1 order of magnitude higher than simultaneous administration in all the variables studied except for SIRT1, whose strong downregulation induced by d-gal was equally prevented by both schedules. The benefits of vitamin D for oxidative stress were stronger in the brain than in the heart. Brain MDA levels were more sensitive to damage, while SOD and GPx antioxidant enzymatic activities were in the heart. In this order, the magnitude of SOD, MDA, and GPx oxidative stress markers was sensitive to prevention. In summary, the results unveiled distinct aging induction, preventive signatures, and sensitivity of markers depending on different levels of study and tissues, which are relevant from a mechanistic view and in the design of targeted interventions.
Collapse
Affiliation(s)
- Sahar Salemi
- Department of Biochemistry, School of MedicineRafsanjan University of Medical SciencesRafsanjanIran
| | - Mohammad Yasin Zamanian
- Neurophysiology Research CenterHamadan University of Medical SciencesHamadanIran
- Department of Pharmacology and Toxicology, School of PharmacyHamadan University of Medical SciencesHamadanIran
| | - Lydia Giménez‐Llort
- Institute of Neuroscience & Department of Psychiatry and Forensic MedicineUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Zahra Jalali
- Department of Biochemistry, School of MedicineRafsanjan University of Medical SciencesRafsanjanIran
| | - Mehdi Mahmoodi
- Department of Clinical Biochemistry, Afzalipoor Faculty of MedicineKerman University of Medical SciencesKermanIran
| | | | - Ayat Kaeidi
- Department of Physiology, School of MedicineRafsanjan University of Medical SciencesRafsanjanIran
| | - Zahra Taghipour
- Department of Anatomy, School of MedicineRafsanjan University of Medical SciencesRafsanjanIran
| | - Morteza Khademalhosseini
- Department of Pathology, School of MedicineRafsanjan University of Medical SciencesRafsanjanIran
| | - Mona Modanloo
- Pharmaceutical Sciences Research CenterMazandaran University of Medical SciencesSariIran
| | - Mohammad Reza Hajizadehi
- Department of Biochemistry, School of MedicineRafsanjan University of Medical SciencesRafsanjanIran
| |
Collapse
|
50
|
Yang Y, Zhang X, Li D, Fang R, Wang Z, Yun D, Wang M, Wang J, Dong H, Fei Z, Li Q, Liu Z, Shen C, Fei J, Yu M, Behnisch T, Huang F. NRSF regulates age-dependently cognitive ability and its conditional knockout in APP/PS1 mice moderately alters AD-like pathology. Hum Mol Genet 2023; 32:2558-2575. [PMID: 36229920 DOI: 10.1093/hmg/ddac253] [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: 06/18/2022] [Revised: 09/19/2022] [Accepted: 10/06/2022] [Indexed: 11/12/2022] Open
Abstract
NRSF/REST (neuron-restrictive silencer element, also known as repressor element 1-silencing transcription factor), plays a key role in neuronal homeostasis as a transcriptional repressor of neuronal genes. NRSF/REST relates to cognitive preservation and longevity of humans, but its specific functions in age-dependent and Alzheimer's disease (AD)-related memory deficits remain unclear. Here, we show that conditional NRSF/REST knockout either in the dorsal telencephalon or specially in neurons induced an age-dependently diminished retrieval performance in spatial or fear conditioning memory tasks and altered hippocampal synaptic transmission and activity-dependent synaptic plasticity. The NRSF/REST deficient mice were also characterized by an increase of activated glial cells, complement C3 protein and the transcription factor C/EBPβ in the cortex and hippocampus. Reduction of NRSF/REST by conditional depletion upregulated the activation of astrocytes in APP/PS1 mice, and increased the C3-positive glial cells, but did not alter the Aβ loads and memory retrieval performances of 6- and 12-month-old APP/PS1 mice. Simultaneously, overexpression of NRSF/REST improved cognitive abilities of aged wild type, but not in AD mice. These findings demonstrated that NRSF/REST is essential for the preservation of memory performance and activity-dependent synaptic plasticity during aging and takes potential roles in the onset of age-related memory impairments. However, while altering the glial activation, NRSF/REST deficiency does not interfere with the Aβ deposits and the electrophysiological and cognitive AD-like pathologies.
Collapse
Affiliation(s)
- Yufang Yang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Xiaoshuang Zhang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Dongxue Li
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
- Department of Endocrinology and Metabolism, School of Medicine, Shanghai Tenth People's Hospital of Tongji University, No. 301 Middle Yanchang Road, Shanghai 200072, China
| | - Rong Fang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Zishan Wang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Di Yun
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Mo Wang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Jinghui Wang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Hongtian Dong
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Zhaoliang Fei
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University of Medicine, Shanghai 200240, China
| | - Qing Li
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Zhaolin Liu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Chenye Shen
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Jian Fei
- Shanghai Engineering Research Center for Model Organisms, Shanghai Model Organisms Center, INC., Shanghai 201203, China
- School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Mei Yu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Thomas Behnisch
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Fang Huang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| |
Collapse
|