1
|
Han M, Zeng D, Tan W, Chen X, Bai S, Wu Q, Chen Y, Wei Z, Mei Y, Zeng Y. Brain region-specific roles of brain-derived neurotrophic factor in social stress-induced depressive-like behavior. Neural Regen Res 2025; 20:159-173. [PMID: 38767484 PMCID: PMC11246125 DOI: 10.4103/nrr.nrr-d-23-01419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 01/19/2024] [Indexed: 05/22/2024] Open
Abstract
Brain-derived neurotrophic factor is a key factor in stress adaptation and avoidance of a social stress behavioral response. Recent studies have shown that brain-derived neurotrophic factor expression in stressed mice is brain region-specific, particularly involving the corticolimbic system, including the ventral tegmental area, nucleus accumbens, prefrontal cortex, amygdala, and hippocampus. Determining how brain-derived neurotrophic factor participates in stress processing in different brain regions will deepen our understanding of social stress psychopathology. In this review, we discuss the expression and regulation of brain-derived neurotrophic factor in stress-sensitive brain regions closely related to the pathophysiology of depression. We focused on associated molecular pathways and neural circuits, with special attention to the brain-derived neurotrophic factor-tropomyosin receptor kinase B signaling pathway and the ventral tegmental area-nucleus accumbens dopamine circuit. We determined that stress-induced alterations in brain-derived neurotrophic factor levels are likely related to the nature, severity, and duration of stress, especially in the above-mentioned brain regions of the corticolimbic system. Therefore, BDNF might be a biological indicator regulating stress-related processes in various brain regions.
Collapse
Affiliation(s)
- Man Han
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Deyang Zeng
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Wei Tan
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Xingxing Chen
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Shuyuan Bai
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Qiong Wu
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Yushan Chen
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Zhen Wei
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Yufei Mei
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Yan Zeng
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
2
|
Boyzo Montes de Oca A, Tendilla-Beltrán H, Bringas ME, Flores G, Aceves J. Chronic pramipexole and rasagiline treatment enhances dendritic spine structural neuroplasticity in striatal and prefrontal cortex neurons of rats with bilateral intrastriatal 6-hydroxydopamine lesions. J Chem Neuroanat 2024; 141:102468. [PMID: 39383978 DOI: 10.1016/j.jchemneu.2024.102468] [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: 07/22/2024] [Revised: 09/26/2024] [Accepted: 10/06/2024] [Indexed: 10/11/2024]
Abstract
Parkinson's disease manifests as neurological alterations within dendritic spines in the striatal and neocortical brain regions, where their functionality closely correlates with morphology. However, the impact of current pharmacotherapy on dendritic spine neuroplasticity, crucial for novel drug development in neurological and psychiatric disorders, remains unclear. This study investigated the effects of 6-OHDA intrastriatal bilateral lesions in male adult rats on behavior and dendritic spine neuroplasticity in striatal and cortical neurons. Furthermore, it evaluated the influence of chronic co-administration of pramipexole (PPX), a D3 receptor agonist, and rasagiline (Ras), a selective MAO-B inhibitor, on these alterations. Lesioned animals exhibited impaired balance behavior, with no improvement following PPX-Ras treatment. The 6-OHDA lesion decreased dendritic spine density in caudate putamen (CPU) spiny projection neurons (SPNs), a change unaffected by treatment, though PPX-Ras increased mushroom spines and reduced stubby spines in these neurons. In nucleus accumbens (NAcc) SPNs and prefrontal cortex layer 3 (PFC-3) pyramidal cells, dendritic spine density remained unaltered, but PPX-Ras decreased mushroom spines and increased bifurcated spines in the NAcc, while increasing mushroom spines and decreasing stubby spines in PFC-3 in lesioned rats. These findings emphasize the importance of dendritic spines as promising targets for innovative pharmacotherapies for Parkinson's disease.
Collapse
Affiliation(s)
- Alfonso Boyzo Montes de Oca
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Mexico City, Mexico
| | - Hiram Tendilla-Beltrán
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - María E Bringas
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Gonzalo Flores
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico.
| | - Jorge Aceves
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Mexico City, Mexico.
| |
Collapse
|
3
|
Ghaedi GH, Nasiri L, Hassanpour H, Mehdi Naghizadeh M, Abdollahzadeh A, Ghazanfari T. Evaluation of serum BDNF, IL-1β, and IL-6 levels alongside assessing mental health and life satisfaction in sulfur mustard-chemical veterans. Int Immunopharmacol 2024; 143:113479. [PMID: 39481191 DOI: 10.1016/j.intimp.2024.113479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/02/2024]
Abstract
Sulfur mustard (SM), a chemical warfare weapon has been used in conflicts. The delayed impact of sulfur mustard on mental and physical health of veterans remains a topic of significant concern. This cross-sectional study investigated the serum levels of brain-derived neurotrophic factor (BDNF), interleukin (IL)-1β, and IL-6 in 227 SM-chemical veterans receiving long-term financial support and 77 healthy individuals. Their mental health status and life satisfaction were assessed through three self-report questionnaires (General Health Questionnaire - 28, GHQ-28; Depression, Anxiety & Stress Scale, DASS-21; 36-Item Short Form Survey, SF-36). Our findings revealed higher levels of anxiety/insomnia, and psychiatric symptoms in the veterans compared to the control group (P < 0.05), accompanied by depression, stress, and anxiety as measured by the GHQ-28 and DASS-21 assessments. Severe depression and social dysfunction were not prevalent in the veterans compared to the control group (P > 0.05) according to the GHQ-28 findings. The SF-36 assessment indicated overall better health conditions for SM participants, with higher scores across various domains (general health, social function, and mental health) and two mental and physical dimensions in the veterans compared to the control group (P < 0.05). IL-1β and IL-6 levels were lower in the SM-exposed group than in the control group, while the BDNF level was higher in the SM-exposed group (P < 0.05). Alterations in BDNF, IL-1β, and IL-6 levels along with results of the mentioned questionnaires may be evidence of partial improvement in the mental and physical health of the SM-exposed individuals receiving the financial support.
Collapse
Affiliation(s)
| | - Leila Nasiri
- Health Equity Research Center, Shahed University, Tehran, Iran
| | - Hossein Hassanpour
- Health Equity Research Center, Shahed University, Tehran, Iran; Department of Basic Sciences, Faculty of Veterinary Medicine, Shahrekord University, Shahrekord, Iran
| | | | - Ahmad Abdollahzadeh
- Chemical Victims' Clinic of Sardasht, Sardasht, West Azerbaijan, Iran; Immunoregulation Research Center, Shahed University, Tehran, Iran
| | - Tooba Ghazanfari
- Immunoregulation Research Center, Shahed University, Tehran, Iran.
| |
Collapse
|
4
|
Pinkston BTC, Browning JL, Olsen ML. Astrocyte TrkB.T1 deficiency disrupts glutamatergic synaptogenesis and astrocyte-synapse interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619696. [PMID: 39484608 PMCID: PMC11526899 DOI: 10.1101/2024.10.22.619696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Perisynaptic astrocyte processes (PAPs) contact pre- and post-synaptic elements to provide structural and functional support to synapses. Accumulating research demonstrates that the cradling of synapses by PAPs is critical for synapse formation, stabilization, and plasticity. The specific signaling pathways that govern these astrocyte-synapse interactions, however, remain to be elucidated. Herein, we demonstrate a role for the astrocyte TrkB.T1 receptor, a truncated isoform of the canonical receptor for brain derived neurotrophic factor (BDNF), in modulating astrocyte-synapse interactions and excitatory synapse development. Neuron-astrocyte co-culture studies revealed that loss of astrocyte TrkB.T1 disrupts the formation of PAPs. To elucidate the role of TrkB.T1 in synapse development, we conditionally deleted TrkB.T1 in astrocytes in mice. Synaptosome preparations were employed to probe for TrkB.T1 localization at the PAP, and confocal three-dimensional microscopy revealed a significant reduction in synapse density and astrocyte-synapse interactions across development in the absence of astrocytic TrkB.T1. These findings suggest that BDNF/TrkB.T1 signaling in astrocytes is critical for normal excitatory synapse formation in the cortex and that astrocyte TrkB.T1 serves a requisite role in astrocyte synapse interactions. Overall, this work provides new insights into the molecular mechanisms of astrocyte-mediated synaptogenesis and may have implications for understanding neurodevelopmental disorders and developing potential therapeutic targets.
Collapse
|
5
|
Naseem M, Khan H, Parvez S. TrkB-BDNF Signalling and Arc/Arg3.1 Immediate Early Genes in the Anterior Cingulate Cortex and Hippocampus: Insights into Novel Memory Milestones Through Behavioural Tagging. Mol Neurobiol 2024; 61:8307-8319. [PMID: 38485841 DOI: 10.1007/s12035-024-04071-1] [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: 10/27/2023] [Accepted: 02/26/2024] [Indexed: 09/21/2024]
Abstract
In recent years, there has been a surge in interest in investigating the mechanisms underlying memory consolidation. However, our understanding of the behavioural tagging (BT) model and its establishment in diverse brain regions remains limited. This study elucidates the contributions of the anterior cingulate cortex (ACC) and hippocampus in the formation of long-term memory (LTM) employing behaviour tagging as a model for studying the underlying mechanism of LTM formation in rats. Existing knowledge highlights a protein synthesis-dependent phase as imperative for LTM. Brain-derived neurotrophic factor (BDNF) stands as a pivotal plasticity-related protein (PRP) in mediating molecular alterations crucial for long-term synaptic plasticity and memory consolidation. Our study offers evidence suggesting that tropomyosin receptor kinase B (TrkB), the receptor of BDNF, may act as a combined "behavioural tag/PRP". Interfering with the expression of these molecules resulted in impaired LTM after 24 h. Furthermore, augmenting BDNF expression led to an elevation in Arc protein levels in both the ACC and hippocampus regions. Introducing novelty around weak inhibitory avoidance (IA) training resulted in heightened step-down latencies and expression of these molecules, respectively. We also demonstrate that the increase in Arc expression relies on BDNF synthesis, which is vital for the memory consolidation process. Additionally, inhibiting BDNF using an anti-BDNF function-blocking antibody impacted Arc expression in both the ACC and hippocampus regions, disrupting the transformations from labile to robust memory. These findings mark the initial identification of a "behavioural tag/PRP" combination and underscore the involvement of the TrkB-BDNF-Arc cascade in the behavioural tagging model of learning and memory.
Collapse
Affiliation(s)
- Mehar Naseem
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Hiba Khan
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
6
|
Fursa GA, Andretsova SS, Shishkina VS, Voronova AD, Karsuntseva EK, Chadin AV, Reshetov IV, Stepanova OV, Chekhonin VP. The Use of Neurotrophic Factors as a Promising Strategy for the Treatment of Neurodegenerative Diseases (Review). Bull Exp Biol Med 2024:10.1007/s10517-024-06218-5. [PMID: 39266924 DOI: 10.1007/s10517-024-06218-5] [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: 08/07/2023] [Indexed: 09/14/2024]
Abstract
The review considers the use of exogenous neurotrophic factors in the treatment of neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, multiple sclerosis, and others. This group of diseases is associated with the death of neurons and dysfunction of the nervous tissue. Currently, there is no effective therapy for neurodegenerative diseases, and their treatment remains a serious problem of modern medicine. A promising strategy is the use of exogenous neurotrophic factors. Targeted delivery of these factors to the nervous tissue can improve survival of neurons during the development of neurodegenerative processes and ensure neuroplasticity. There are methods of direct injection of neurotrophic factors into the nervous tissue, delivery using viral vectors, as well as the use of gene cell products. The effectiveness of these approaches has been studied in numerous experimental works and in a number of clinical trials. Further research in this area could provide the basis for the creation of an alternative treatment for neurodegenerative diseases.
Collapse
Affiliation(s)
- G A Fursa
- V. Serbsky National Medical Research Centre for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia.
- Pirogov Russian National Research Medical University, Moscow, Russia.
- National Medical Research Centre of Cardiology named after academician E. I. Chazov, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - S S Andretsova
- V. Serbsky National Medical Research Centre for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - V S Shishkina
- V. Serbsky National Medical Research Centre for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A D Voronova
- V. Serbsky National Medical Research Centre for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
- National Medical Research Centre of Cardiology named after academician E. I. Chazov, Ministry of Health of the Russian Federation, Moscow, Russia
| | - E K Karsuntseva
- V. Serbsky National Medical Research Centre for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A V Chadin
- V. Serbsky National Medical Research Centre for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - I V Reshetov
- University Clinical Hospital No. 1, I. M. Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
- Academy of Postgraduate Education, Federal Research and Clinical Center of Specialized Types of Health Care and Medical Technology of the Federal Medical and Biological Agency, Moscow, Russia
| | - O V Stepanova
- V. Serbsky National Medical Research Centre for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
- National Medical Research Centre of Cardiology named after academician E. I. Chazov, Ministry of Health of the Russian Federation, Moscow, Russia
| | - V P Chekhonin
- V. Serbsky National Medical Research Centre for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
7
|
Yılmaz E, Baltaci SB, Mogulkoc R, Baltaci AK. The impact of flavonoids and BDNF on neurogenic process in various physiological/pathological conditions including ischemic insults: a narrative review. Nutr Neurosci 2024; 27:1025-1041. [PMID: 38151886 DOI: 10.1080/1028415x.2023.2296165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
OBJECTIVE Ischemic stroke is the leading cause of mortality and disability worldwide with more than half of survivors living with serious neurological sequelae thus, it has recently attracted considerable attention in the field of medical research. Neurogenesis is the process of formation of new neurons in the brain, including the human brain, from neural stem/progenitor cells [NS/PCs] which reside in neurogenic niches that contain the necessary substances for NS/PC proliferation, differentiation, migration, and maturation into functioning neurons which can integrate into a pre-existing neural network.Neurogenesis can be modulated by many exogenous and endogenous factors, pathological conditions. Both brain-derived neurotrophic factor, and flavonoids can modulate the neurogenic process in physiological conditions and after various pathological conditions including ischemic insults. AIM This review aims to discuss neurogenesis after ischemic insults and to determine the role of flavonoids and BDNF on neurogenesis under physiological and pathological conditions with a concentration on ischemic insults to the brain in particular. METHOD Relevant articles assessing the impact of flavonoids and BDNF on neurogenic processes in various physiological/pathological conditions including ischemic insults within the timeline of 1965 until 2023 were searched using the PubMed database. CONCLUSIONS The selected studies have shown that ischemic insults to the brain induce NS/PC proliferation, differentiation, migration, and maturation into functioning neurons integrating into a pre-existing neural network. Flavonoids and BDNF can modulate neurogenesis in the brain in various physiological/pathological conditions including ischemic insults. In conclusion, flavonoids and BDNF may be involved in post-ischemic brain repair processes through enhancing endogenous neurogenesis.
Collapse
Affiliation(s)
- Esen Yılmaz
- Selcuk University, Medical Faculty, Department of Physiology, Konya, Turkey
| | | | - Rasim Mogulkoc
- Selcuk University, Medical Faculty, Department of Physiology, Konya, Turkey
| | | |
Collapse
|
8
|
De Jager JE, Boesjes R, Roelandt GHJ, Koliaki I, Sommer IEC, Schoevers RA, Nuninga JO. Shared effects of electroconvulsive shocks and ketamine on neuroplasticity: A systematic review of animal models of depression. Neurosci Biobehav Rev 2024; 164:105796. [PMID: 38981574 DOI: 10.1016/j.neubiorev.2024.105796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/11/2024]
Abstract
Electroconvulsive shocks (ECS) and ketamine are antidepressant treatments with a relatively fast onset of therapeutic effects compared to conventional medication and psychotherapy. While the exact neurobiological mechanisms underlying the antidepressant response of ECS and ketamine are unknown, both interventions are associated with neuroplasticity. Restoration of neuroplasticity may be a shared mechanism underlying the antidepressant efficacy of these interventions. In this systematic review, literature of animal models of depression is summarized to examine the possible role of neuroplasticity in ECS and ketamine on a molecular, neuronal, synaptic and functional level, and specifically to what extent these mechanisms are shared between both interventions. The results highlight that hippocampal neurogenesis and brain-derived neurotrophic factor (BDNF) levels are consistently increased after ECS and ketamine. Moreover, both interventions positively affect glutamatergic neurotransmission, astrocyte and neuronal morphology, synaptic density, vasculature and functional plasticity. However, a small number of studies investigated these processes after ECS. Understanding the shared fundamental mechanisms of fast-acting antidepressants can contribute to the development of novel therapeutic approaches for patients with severe depression.
Collapse
Affiliation(s)
- Jesca E De Jager
- Department of Biomedical Sciences, Brain Center, University Medical Center, Groningen, the Netherlands.
| | - Rutger Boesjes
- University Centre of Psychiatry, University Medical Center Groningen, the Netherlands
| | - Gijs H J Roelandt
- University Centre of Psychiatry, University Medical Center Groningen, the Netherlands
| | - Ilektra Koliaki
- University Centre of Psychiatry, University Medical Center Groningen, the Netherlands
| | - Iris E C Sommer
- Department of Biomedical Sciences, Brain Center, University Medical Center, Groningen, the Netherlands
| | - Robert A Schoevers
- University Centre of Psychiatry, University Medical Center Groningen, the Netherlands
| | - Jasper O Nuninga
- Department of Biomedical Sciences, Brain Center, University Medical Center, Groningen, the Netherlands; University Medical Centre Utrecht, Department of Psychiatry, the Netherlands
| |
Collapse
|
9
|
Navarro-Lobato I, Masmudi-Martín M, López-Aranda MF, López-Téllez JF, Delgado G, Granados-Durán P, Gaona-Romero C, Carretero-Rey M, Posadas S, Quiros-Ortega ME, Khan ZU. Promotion of structural plasticity in area V2 of visual cortex prevents against object recognition memory deficits in aging and Alzheimer's disease rodents. Neural Regen Res 2024; 19:1835-1841. [PMID: 38103251 PMCID: PMC10960297 DOI: 10.4103/1673-5374.389301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/23/2023] [Accepted: 10/26/2023] [Indexed: 12/18/2023] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202408000-00038/figure1/v/2023-12-16T180322Z/r/image-tiff Memory deficit, which is often associated with aging and many psychiatric, neurological, and neurodegenerative diseases, has been a challenging issue for treatment. Up till now, all potential drug candidates have failed to produce satisfactory effects. Therefore, in the search for a solution, we found that a treatment with the gene corresponding to the RGS14414 protein in visual area V2, a brain area connected with brain circuits of the ventral stream and the medial temporal lobe, which is crucial for object recognition memory (ORM), can induce enhancement of ORM. In this study, we demonstrated that the same treatment with RGS14414 in visual area V2, which is relatively unaffected in neurodegenerative diseases such as Alzheimer's disease, produced long-lasting enhancement of ORM in young animals and prevent ORM deficits in rodent models of aging and Alzheimer's disease. Furthermore, we found that the prevention of memory deficits was mediated through the upregulation of neuronal arborization and spine density, as well as an increase in brain-derived neurotrophic factor (BDNF). A knockdown of BDNF gene in RGS14414-treated aging rats and Alzheimer's disease model mice caused complete loss in the upregulation of neuronal structural plasticity and in the prevention of ORM deficits. These findings suggest that BDNF-mediated neuronal structural plasticity in area V2 is crucial in the prevention of memory deficits in RGS14414-treated rodent models of aging and Alzheimer's disease. Therefore, our findings of RGS14414 gene-mediated activation of neuronal circuits in visual area V2 have therapeutic relevance in the treatment of memory deficits.
Collapse
Affiliation(s)
- Irene Navarro-Lobato
- Laboratory of Neurobiology, Centro de Investigaciones Médico Sanitarias (CIMES), University of Malaga, Malaga, Spain
- Department of Medicine, Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Mariam Masmudi-Martín
- Laboratory of Neurobiology, Centro de Investigaciones Médico Sanitarias (CIMES), University of Malaga, Malaga, Spain
- Department of Medicine, Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Manuel F. López-Aranda
- Laboratory of Neurobiology, Centro de Investigaciones Médico Sanitarias (CIMES), University of Malaga, Malaga, Spain
- Department of Medicine, Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Juan F. López-Téllez
- Laboratory of Neurobiology, Centro de Investigaciones Médico Sanitarias (CIMES), University of Malaga, Malaga, Spain
- Department of Medicine, Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Gloria Delgado
- Laboratory of Neurobiology, Centro de Investigaciones Médico Sanitarias (CIMES), University of Malaga, Malaga, Spain
- Department of Medicine, Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Pablo Granados-Durán
- Laboratory of Neurobiology, Centro de Investigaciones Médico Sanitarias (CIMES), University of Malaga, Malaga, Spain
- Department of Medicine, Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Celia Gaona-Romero
- Laboratory of Neurobiology, Centro de Investigaciones Médico Sanitarias (CIMES), University of Malaga, Malaga, Spain
- Department of Medicine, Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Marta Carretero-Rey
- Laboratory of Neurobiology, Centro de Investigaciones Médico Sanitarias (CIMES), University of Malaga, Malaga, Spain
- Department of Medicine, Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Sinforiano Posadas
- Laboratory of Neurobiology, Centro de Investigaciones Médico Sanitarias (CIMES), University of Malaga, Malaga, Spain
- Department of Medicine, Faculty of Medicine, University of Malaga, Malaga, Spain
| | - María E. Quiros-Ortega
- Laboratory of Neurobiology, Centro de Investigaciones Médico Sanitarias (CIMES), University of Malaga, Malaga, Spain
- Department of Medicine, Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Zafar U. Khan
- Laboratory of Neurobiology, Centro de Investigaciones Médico Sanitarias (CIMES), University of Malaga, Malaga, Spain
- Department of Medicine, Faculty of Medicine, University of Malaga, Malaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
10
|
Zhang Z, Wu H, Wang S, Li Y, Yang P, Xu L, Liu Y, Liu M. PRG ameliorates cognitive impairment in Alzheimer's disease mice by regulating β-amyloid and targeting the ERK pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155671. [PMID: 38763005 DOI: 10.1016/j.phymed.2024.155671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/13/2024] [Accepted: 04/21/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND PRG is derived from Phellinus ribis and is a homogeneous polysaccharide with well-defined structural information. PRG was found to have significant in vitro neurotrophic and neuroprotective activities. Thus, PRG might be a potential treatment for Alzheimer's disease. However, the related mechanisms of action are still unclear, so deeper in vivo experimental validation and the potential mechanisms need to be investigated. PURPOSE The effects of PRG on AD mice were investigated using Senescence-accelerated SAMP8 mice as an AD model to elucidate the crucial molecular mechanisms. METHODS PRG was obtained from Phellinus ribis by water-alcohol precipitation, column chromatography, and ultrafiltration. The Morris water maze and novel object recognition behavioral assays were used to evaluate the effects of PRG in AD mice. Nissl staining, the TUNEL apoptosis assay, and Golgi staining were used to assess brain neuronal cell damage, apoptosis, and neuronal status. Enzyme-linked immunosorbent assays, Western blotting, and immunofluorescence were used to explore the impacts of correlated factors and protein pathways under relevant mechanisms. RESULTS The findings suggest that PRG improved learning ability and spatial memory capacity in SAMP8 mice. PRG hastened the disintegration of β-amyloid, reduced the content and abnormal accumulation of the toxic Aβ1-42 protein, and decreased apoptosis. PRG activated the BDNF/ERK/CREB signaling pathway through a cascade, exerted neurotrophic effects, regulated cell proliferation and differentiation, increased neuronal dendritic branching and spine density, and improved synaptic plasticity. CONCLUSION PRG promoted β-amyloid degradation to reduce neuronal damage and apoptosis. It exerted neurotrophic effects by activating the BDNF/ERK/CREB pathway, promoting neuronal dendritic branching and dendritic spine growth, regulating cell proliferation and differentiation, and improving synaptic plasticity, which improved AD. Taken together, as a novel natural active polysaccharide with a well-defined structure, PRG affected AD symptoms in senescence-accelerated mice by interacting with multiple targets. The results indicate that PRG is a promising potential anti-AD drug candidate.
Collapse
Affiliation(s)
- Zhiyuan Zhang
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Haoran Wu
- School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Shuai Wang
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yuanyuan Li
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Pei Yang
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Lingchuan Xu
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yuhong Liu
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Maoxuan Liu
- Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|
11
|
Lu MN, Wang D, Ye CJ, Yan GJ, Song JF, Shi XY, Li SS, Liu LN, Zhang HX, Dong XH, Hu T, Wang XY, Xiyang YB. Navβ2 Intracellular Fragments Contribute to Aβ1-42-Induced Cognitive Impairment and Synaptic Deficit Through Transcriptional Suppression of BDNF. Mol Neurobiol 2024:10.1007/s12035-024-04317-y. [PMID: 38965172 DOI: 10.1007/s12035-024-04317-y] [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: 02/17/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024]
Abstract
A pathological hallmark of Alzheimer's disease (AD) is the region-specific accumulation of the amyloid-beta protein (Aβ), which triggers aberrant neuronal excitability, synaptic impairment, and progressive cognitive decline. Previous works have demonstrated that Aβ pathology induced aberrant elevation in the levels and excessive enzymatic hydrolysis of voltage-gated sodium channel type 2 beta subunit (Navβ2) in the brain of AD models, accompanied by alteration in excitability of hippocampal neurons, synaptic deficits, and subsequently, cognitive dysfunction. However, the mechanism is unclear. In this research, by employing cell models treated with toxic Aβ1-42 and AD mice, the possible effects and potential mechanisms induced by Navβ2. The results reveal that Aβ1-42 induces remarkable increases in Navβ2 intracellular domain (Navβ2-ICD) and decreases in both BDNF exons and protein levels, as well as phosphorylated tropomyosin-related kinase B (pTrkB) expression in cells and mice, coupled with cognitive impairments, synaptic deficits, and aberrant neuronal excitability. Administration with exogenous Navβ2-ICD further enhances these effects induced by Aβ1-42, while interfering the generation of Navβ2-ICD and/or complementing BDNF neutralize the Navβ2-ICD-conducted effects. Luciferase reporter assay verifies that Navβ2-ICD regulates BDNF transcription and expression by targeting its promoter. Collectively, our findings partially elucidate that abnormal enzymatic hydrolysis of Navβ2 induced by Aβ1-42-associated AD pathology leads to intracellular Navβ2-ICD overload, which may responsible to abnormal neuronal excitability, synaptic deficit, and cognition dysfunction, through its transcriptional suppression on BDNF. Therefore, this work supplies novel evidences that Navβ2 plays crucial roles in the occurrence and progression of cognitive impairment of AD by transcriptional regulatory activity of its cleaved ICD.
Collapse
Affiliation(s)
- Min-Nan Lu
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
- Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Dan Wang
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Chen-Jun Ye
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Guo-Ji Yan
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Jing-Feng Song
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Xin-Ying Shi
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Shan-Shan Li
- Experimental Teaching Center, Basic Medical College, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Li-Na Liu
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Hui-Xiang Zhang
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Xiao-Han Dong
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Tao Hu
- Department of Laboratory Medicine, The Third People's Hospital of Yunnan Province, Kunming, 650000, Yunnan, China
| | - Xu-Yang Wang
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yan-Bin Xiyang
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China.
| |
Collapse
|
12
|
Mey M, Bhatta S, Suresh S, Labrador LM, Piontkivska H, Casadesus G. Therapeutic benefits of central LH receptor agonism in the APP/PS1 AD model involve trophic and immune regulation and are reproductive status dependent. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167165. [PMID: 38653355 DOI: 10.1016/j.bbadis.2024.167165] [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: 02/06/2024] [Revised: 04/02/2024] [Accepted: 04/08/2024] [Indexed: 04/25/2024]
Abstract
The mechanisms that underly reproductive hormone effects on cognition, neuronal plasticity, and AD risk, particularly in relation to gonadotropin LH receptor (LHCGR) signaling, remain poorly understood. To address this gap in knowledge and clarify the impact of circulating steroid hormones on the therapeutic effects of CNS LHCGR activation, we delivered the LHCGR agonist human chorionic gonadotropin (hCG) intracerebroventricularly (ICV) and evaluated functional, structural, plasticity-related signaling cascades, Aβ pathology, and transcriptome differences in reproductively intact and ovariectomized (OVX) APP/PS1 AD female mice. Here we demonstrate that CNS hCG delivery restored function to wild-type levels only in OVX APP/PS1 mice. Spine density was increased in all hCG treated groups independently of reproductive status. Notably, increases in BDNF signaling and cognition, were selectively upregulated only in the OVX hCG-treated group. RNA sequencing analyses identified a significant increase in peripheral myeloid and pro-inflammatory genes within the hippocampi of the OVX group that were completely reversed by hCG treatment, identifying a potential mechanism underlying the selective therapeutic effect of LHCGR activation. Interestingly, in intact mice, hCG administration mimicked the effects of gonadectomy. Together, our findings indicate that CNS LHCGR agonism in the post-menopausal context is beneficial through trophic and immune mechanisms. Our findings also underscore the presence of a steroid-LHCGR mechanistic interaction that is unexplored yet potentially meaningful to fully understand "post-menopausal" brain function and CNS hormone treatment response.
Collapse
Affiliation(s)
- Megan Mey
- Kent State University, Kent, OH 44240, United States of America
| | - Sabina Bhatta
- Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Sneha Suresh
- University of Florida, Gainesville, FL 32606, United States of America
| | | | | | - Gemma Casadesus
- University of Florida, Gainesville, FL 32606, United States of America.
| |
Collapse
|
13
|
Davis N, Taylor B, Abelleira-Hervas L, Karimian-Marnani N, Aleksynas R, Syed N, Di Giovanni S, Palmisano I, Sastre M. Histone deacetylase-3 regulates the expression of the amyloid precursor protein and its inhibition promotes neuroregenerative pathways in Alzheimer's disease models. FASEB J 2024; 38:e23659. [PMID: 38733301 DOI: 10.1096/fj.202301762rr] [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: 04/04/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024]
Abstract
HDAC3 inhibition has been shown to improve memory and reduce amyloid-β (Aβ) in Alzheimer's disease (AD) models, but the underlying mechanisms are unclear. We investigated the molecular effects of HDAC3 inhibition on AD pathology, using in vitro and ex vivo models of AD, based on our finding that HDAC3 expression is increased in AD brains. For this purpose, N2a mouse neuroblastoma cells as well as organotypic brain cultures (OBCSs) of 5XFAD and wild-type mice were incubated with various concentrations of the HDAC3 selective inhibitor RGFP966 (0.1-10 μM) for 24 h. Treatment with RGFP966 or HDAC3 knockdown in N2a cells was associated with an increase on amyloid precursor protein (APP) and mRNA expressions, without alterations in Aβ42 secretion. In vitro chromatin immunoprecipitation analysis revealed enriched HDAC3 binding at APP promoter regions. The increase in APP expression was also detected in OBCSs from 5XFAD mice incubated with 1 μM RGFP966, without changes in Aβ. In addition, HDAC3 inhibition resulted in a reduction of activated Iba-1-positive microglia and astrocytes in 5XFAD slices, which was not observed in OBCSs from wild-type mice. mRNA sequencing analysis revealed that HDAC3 inhibition modulated neuronal regenerative pathways related to neurogenesis, differentiation, axonogenesis, and dendritic spine density in OBCSs. Our findings highlight the complexity and diversity of the effects of HDAC3 inhibition on AD models and suggest that HDAC3 may have multiple roles in the regulation of APP expression and processing, as well as in the modulation of neuroinflammatory and neuroprotective genes.
Collapse
Affiliation(s)
- Nicola Davis
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Ben Taylor
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | | | | | - Robertas Aleksynas
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Nelofer Syed
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Simone Di Giovanni
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Ilaria Palmisano
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
- Department of Neuroscience, The Ohio State University, Columbus, Ohio, USA
- Department of Plastic and Reconstructive Surgery, The Ohio State University, Columbus, Ohio, USA
| | - Magdalena Sastre
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| |
Collapse
|
14
|
Afshari M, Gharibzadeh S, Pouretemad H, Roghani M. Reversing valproic acid-induced autism-like behaviors through a combination of low-frequency repeated transcranial magnetic stimulation and superparamagnetic iron oxide nanoparticles. Sci Rep 2024; 14:8082. [PMID: 38582936 PMCID: PMC10998842 DOI: 10.1038/s41598-024-58871-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024] Open
Abstract
Transcranial magnetic stimulation (TMS) is a neurostimulation device used to modulate brain cortex activity. Our objective was to enhance the therapeutic effectiveness of low-frequency repeated TMS (LF-rTMS) in a rat model of autism spectrum disorder (ASD) induced by prenatal valproic acid (VPA) exposure through the injection of superparamagnetic iron oxide nanoparticles (SPIONs). For the induction of ASD, we administered prenatal VPA (600 mg/kg, I.P.) on the 12.5th day of pregnancy. At postnatal day 30, SPIONs were injected directly into the lateral ventricle of the brain. Subsequently, LF-rTMS treatment was applied for 14 consecutive days. Following the treatment period, behavioral analyses were conducted. At postnatal day 60, brain tissue was extracted, and both biochemical and histological analyses were performed. Our data revealed that prenatal VPA exposure led to behavioral alterations, including changes in social interactions, increased anxiety, and repetitive behavior, along with dysfunction in stress coping strategies. Additionally, we observed reduced levels of SYN, MAP2, and BDNF. These changes were accompanied by a decrease in dendritic spine density in the hippocampal CA1 area. However, LF-rTMS treatment combined with SPIONs successfully reversed these dysfunctions at the behavioral, biochemical, and histological levels, introducing a successful approach for the treatment of ASD.
Collapse
Affiliation(s)
- Masoud Afshari
- Department of Cognitive Psychology, Institute for Cognitive and Brain Sciences, Shahid Beheshti University, Tehran, Iran
| | - Shahriar Gharibzadeh
- Department of Cognitive Psychology, Institute for Cognitive and Brain Sciences, Shahid Beheshti University, Tehran, Iran.
| | - Hamidreza Pouretemad
- Department of Cognitive Psychology, Institute for Cognitive and Brain Sciences, Shahid Beheshti University, Tehran, Iran
| | - Mehrdad Roghani
- Neurophysiology Research Center, Shahed University, Tehran, Iran.
| |
Collapse
|
15
|
Ichimura-Shimizu M, Kurrey K, Miyata M, Dezawa T, Tsuneyama K, Kojima M. Emerging Insights into the Role of BDNF on Health and Disease in Periphery. Biomolecules 2024; 14:444. [PMID: 38672461 PMCID: PMC11048455 DOI: 10.3390/biom14040444] [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: 12/28/2023] [Revised: 03/06/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a growth factor that promotes the survival and growth of developing neurons. It also enhances circuit formation to synaptic transmission for mature neurons in the brain. However, reduced BDNF expression and single nucleotide polymorphisms (SNP) are reported to be associated with functional deficit and disease development in the brain, suggesting that BDNF is a crucial molecule for brain health. Interestingly, BDNF is also expressed in the hypothalamus in appetite and energy metabolism. Previous reports demonstrated that BDNF knockout mice exhibited overeating and obesity phenotypes remarkably. Therefore, we could raise a hypothesis that the loss of function of BDNF may be associated with metabolic syndrome and peripheral diseases. In this review, we describe our recent finding that BDNF knockout mice develop metabolic dysfunction-associated steatohepatitis and recent reports demonstrating the role of one of the BDNF receptors, TrkB-T1, in some peripheral organ functions and diseases, and would provide an insight into the role of BDNF beyond the brain.
Collapse
Affiliation(s)
- Mayuko Ichimura-Shimizu
- Department of Pathology and Laboratory Medicine, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan; (M.I.-S.); (K.T.)
| | - Khuleshwari Kurrey
- Department of Neuroscience, School of Medicine, Yale University, New Haven, CT 06520, USA;
| | - Misaki Miyata
- Department of Applied Bioscience, College of Bioscience and Chemistry, Kanazawa Institute of Technology, 3-1 Yatsukaho, Hakusan 924-0838, Japan; (M.M.); (T.D.)
| | - Takuya Dezawa
- Department of Applied Bioscience, College of Bioscience and Chemistry, Kanazawa Institute of Technology, 3-1 Yatsukaho, Hakusan 924-0838, Japan; (M.M.); (T.D.)
| | - Koichi Tsuneyama
- Department of Pathology and Laboratory Medicine, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan; (M.I.-S.); (K.T.)
| | - Masami Kojima
- Department of Applied Bioscience, College of Bioscience and Chemistry, Kanazawa Institute of Technology, 3-1 Yatsukaho, Hakusan 924-0838, Japan; (M.M.); (T.D.)
| |
Collapse
|
16
|
Wood JA, Chaparala S, Bantang C, Chattopadhyay A, Wesesky MA, Kinchington PR, Nimgaonkar VL, Bloom DC, D'Aiuto L. RNA-Seq time-course analysis of neural precursor cell transcriptome in response to herpes simplex Virus-1 infection. J Neurovirol 2024; 30:131-145. [PMID: 38478163 PMCID: PMC11371869 DOI: 10.1007/s13365-024-01198-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/12/2024] [Accepted: 02/20/2024] [Indexed: 09/04/2024]
Abstract
The neurogenic niches within the central nervous system serve as essential reservoirs for neural precursor cells (NPCs), playing a crucial role in neurogenesis. However, these NPCs are particularly vulnerable to infection by the herpes simplex virus 1 (HSV-1). In the present study, we investigated the changes in the transcriptome of NPCs in response to HSV-1 infection using bulk RNA-Seq, compared to those of uninfected samples, at different time points post infection and in the presence or absence of antivirals. The results showed that NPCs upon HSV-1 infection undergo a significant dysregulation of genes playing a crucial role in aspects of neurogenesis, including genes affecting NPC proliferation, migration, and differentiation. Our analysis revealed that the CREB signaling, which plays a crucial role in the regulation of neurogenesis and memory consolidation, was the most consistantly downregulated pathway, even in the presence of antivirals. Additionally, cholesterol biosynthesis was significantly downregulated in HSV-1-infected NPCs. The findings from this study, for the first time, offer insights into the intricate molecular mechanisms that underlie the neurogenesis impairment associated with HSV-1 infection.
Collapse
Affiliation(s)
- Joel A Wood
- Western Psychiatric Institute and Clinic, Department of Psychiatry, University of Pittsburgh School of Medicine, 3811 O'Hara Street, 15213, Pittsburgh, PA, USA
| | - Srilakshmi Chaparala
- Molecular Biology Information Service, Health Sciences Library System / Falk Library, University of Pittsburgh, M722 Alan Magee Scaife Hall / 3550 Terrace Street, 15261, Pittsburgh, PA, USA
| | - Cecilia Bantang
- Western Psychiatric Institute and Clinic, Department of Psychiatry, University of Pittsburgh School of Medicine, 3811 O'Hara Street, 15213, Pittsburgh, PA, USA
| | - Ansuman Chattopadhyay
- Molecular Biology Information Service, Health Sciences Library System / Falk Library, University of Pittsburgh, M722 Alan Magee Scaife Hall / 3550 Terrace Street, 15261, Pittsburgh, PA, USA
| | - Maribeth A Wesesky
- Western Psychiatric Institute and Clinic, Department of Psychiatry, University of Pittsburgh School of Medicine, 3811 O'Hara Street, 15213, Pittsburgh, PA, USA
| | - Paul R Kinchington
- Department of Ophthalmology, University of Pittsburgh, Suite 820, Eye & Ear Building, 203 Lothrop Street, 15213, Pittsburgh, PA, USA
| | - Vishwajit L Nimgaonkar
- Western Psychiatric Institute and Clinic, Department of Psychiatry, University of Pittsburgh School of Medicine, 3811 O'Hara Street, 15213, Pittsburgh, PA, USA
- VA Pittsburgh Healthcare system at U.S. Department of Veterans Affairs, Pittsburgh, PA, USA
| | - David C Bloom
- Academic Research Building, Department of Molecular Genetics and Microbiology, University of Florida, 1200 Newell Drive, R2-231, 32610, Gainesville, FL, USA
| | - Leonardo D'Aiuto
- Western Psychiatric Institute and Clinic, Department of Psychiatry, University of Pittsburgh School of Medicine, 3811 O'Hara Street, 15213, Pittsburgh, PA, USA.
| |
Collapse
|
17
|
Jiang Y. A theory of the neural mechanisms underlying negative cognitive bias in major depression. Front Psychiatry 2024; 15:1348474. [PMID: 38532986 PMCID: PMC10963437 DOI: 10.3389/fpsyt.2024.1348474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 02/16/2024] [Indexed: 03/28/2024] Open
Abstract
The widely acknowledged cognitive theory of depression, developed by Aaron Beck, focused on biased information processing that emphasizes the negative aspects of affective and conceptual information. Current attempts to discover the neurological mechanism underlying such cognitive and affective bias have successfully identified various brain regions associated with severally biased functions such as emotion, attention, rumination, and inhibition control. However, the neurobiological mechanisms of how individuals in depression develop this selective processing toward negative is still under question. This paper introduces a neurological framework centered around the frontal-limbic circuit, specifically analyzing and synthesizing the activity and functional connectivity within the amygdala, hippocampus, and medial prefrontal cortex. Firstly, a possible explanation of how the positive feedback loop contributes to the persistent hyperactivity of the amygdala in depression at an automatic level is established. Building upon this, two hypotheses are presented: hypothesis 1 revolves around the bidirectional amygdalohippocampal projection facilitating the amplification of negative emotions and memories while concurrently contributing to the impediment of the retrieval of opposing information in the hippocampus attractor network. Hypothesis 2 highlights the involvement of the ventromedial prefrontal cortex in the establishment of a negative cognitive framework through the generalization of conceptual and emotional information in conjunction with the amygdala and hippocampus. The primary objective of this study is to improve and complement existing pathological models of depression, pushing the frontiers of current understanding in neuroscience of affective disorders, and eventually contributing to successful recovery from the debilitating affective disorders.
Collapse
Affiliation(s)
- Yuyue Jiang
- University of California, Santa Barbara, Santa Barbara, CA, United States
| |
Collapse
|
18
|
Bimbi G, Tongiorgi E. Chemical LTP induces confinement of BDNF mRNA under dendritic spines and BDNF protein accumulation inside the spines. Front Mol Neurosci 2024; 17:1348445. [PMID: 38450041 PMCID: PMC10914971 DOI: 10.3389/fnmol.2024.1348445] [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: 12/02/2023] [Accepted: 02/05/2024] [Indexed: 03/08/2024] Open
Abstract
The neurotrophin brain-derived neurotrophic factor (BDNF) plays a key role in neuronal development and synaptic plasticity. The discovery that BDNF mRNA can be transported in neuronal dendrites in an activity-dependent manner has suggested that its local translation may support synapse maturation and plasticity. However, a clear demonstration that BDNF mRNA is locally transported and translated at activated synapses in response to long-term potentiation (LTP) is still lacking. Here, we study the dynamics of BDNF mRNA dendritic trafficking following the induction of chemical LTP (cLTP). Dendritic transport of BDNF transcripts was analyzed using the MS2 system for mRNA visualization, and chimeric BDNF-GFP constructs were used to monitor protein synthesis in living neurons. We found that within 15 min from cLTP induction, most BDNF mRNA granules become stationary and transiently accumulate in the dendritic shaft at the base of the dendritic spines, while at 30 min they accumulate inside the spine, similar to the control CamkIIα mRNA which also increased inside the spines at 60 min post-cLTP. At 60 min but not at 15 min from cLTP induction, we observed an increase in BDNF protein levels within the spines. Taken together, these findings suggest that BDNF mRNA trafficking is arrested in the early phase of cLTP, providing a local source of mRNA for BDNF translation at the base of the spine followed by translocation of both the BDNF mRNA and protein within the spine head in the late phase of LTP.
Collapse
Affiliation(s)
| | - Enrico Tongiorgi
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
19
|
Adonina S, Bazhenova E, Bazovkina D. Effect of Short Photoperiod on Behavior and Brain Plasticity in Mice Differing in Predisposition to Catalepsy: The Role of BDNF and Serotonin System. Int J Mol Sci 2024; 25:2469. [PMID: 38473717 DOI: 10.3390/ijms25052469] [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: 12/31/2023] [Revised: 02/14/2024] [Accepted: 02/18/2024] [Indexed: 03/14/2024] Open
Abstract
Seasonal affective disorder is characterized by depression during fall/winter as a result of shorter daylight. Catalepsy is a syndrome of some grave mental diseases. Both the neurotransmitter serotonin (5-HT) and brain-derived neurotrophic factor (BDNF) are involved in the pathophysiological mechanisms underlying catalepsy and depressive disorders. The aim was to compare the response of behavior and brain plasticity to photoperiod alterations in catalepsy-resistant C57BL/6J and catalepsy-prone CBA/Lac male mice. Mice of both strains were exposed for six weeks to standard-day (14 h light/10 h darkness) or short-day (4 h light/20 h darkness) conditions. Short photoperiod increased depressive-like behavior in both strains. Only treated CBA/Lac mice demonstrated increased cataleptic immobility, decreased brain 5-HT level, and the expression of Tph2 gene encoding the key enzyme for 5-HT biosynthesis. Mice of both strains maintained under short-day conditions, compared to those under standard-day conditions, showed a region-specific decrease in the brain transcription of the Htr1a, Htr4, and Htr7 genes. After a short photoperiod exposure, the mRNA levels of the BDNF-related genes were reduced in CBA/Lac mice and were increased in the C57BL/6J mice. Thus, the predisposition to catalepsy considerably influences the photoperiodic changes in neuroplasticity, wherein both C57BL/6J and CBA/Lac mice can serve as a powerful tool for investigating the link between seasons and mood.
Collapse
Affiliation(s)
- Svetlana Adonina
- Federal Research Center Institute of Cytology and Genetics, Siberian Division of the Russian Academy of Science, Lavrentieva 10, Novosibirsk 630090, Russia
| | - Ekaterina Bazhenova
- Federal Research Center Institute of Cytology and Genetics, Siberian Division of the Russian Academy of Science, Lavrentieva 10, Novosibirsk 630090, Russia
| | - Darya Bazovkina
- Federal Research Center Institute of Cytology and Genetics, Siberian Division of the Russian Academy of Science, Lavrentieva 10, Novosibirsk 630090, Russia
| |
Collapse
|
20
|
Zhu Y, Hui Q, Zhang Z, Fu H, Qin Y, Zhao Q, Li Q, Zhang J, Guo L, He W, Han C. Advancements in the study of synaptic plasticity and mitochondrial autophagy relationship. J Neurosci Res 2024; 102:e25309. [PMID: 38400573 DOI: 10.1002/jnr.25309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024]
Abstract
Synapses serve as the points of communication between neurons, consisting primarily of three components: the presynaptic membrane, synaptic cleft, and postsynaptic membrane. They transmit signals through the release and reception of neurotransmitters. Synaptic plasticity, the ability of synapses to undergo structural and functional changes, is influenced by proteins such as growth-associated proteins, synaptic vesicle proteins, postsynaptic density proteins, and neurotrophic growth factors. Furthermore, maintaining synaptic plasticity consumes more than half of the brain's energy, with a significant portion of this energy originating from ATP generated through mitochondrial energy metabolism. Consequently, the quantity, distribution, transport, and function of mitochondria impact the stability of brain energy metabolism, thereby participating in the regulation of fundamental processes in synaptic plasticity, including neuronal differentiation, neurite outgrowth, synapse formation, and neurotransmitter release. This article provides a comprehensive overview of the proteins associated with presynaptic plasticity, postsynaptic plasticity, and common factors between the two, as well as the relationship between mitochondrial energy metabolism and synaptic plasticity.
Collapse
Affiliation(s)
- Yousong Zhu
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Qinlong Hui
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Zheng Zhang
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Hao Fu
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Yali Qin
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Qiong Zhao
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Qinqing Li
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Junlong Zhang
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
| | - Lei Guo
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
| | - Wenbin He
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
| | - Cheng Han
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| |
Collapse
|
21
|
Mohrmann L, Seebach J, Missler M, Rohlmann A. Distinct Alterations in Dendritic Spine Morphology in the Absence of β-Neurexins. Int J Mol Sci 2024; 25:1285. [PMID: 38279285 PMCID: PMC10817056 DOI: 10.3390/ijms25021285] [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: 12/21/2023] [Revised: 01/12/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
Dendritic spines are essential for synaptic function because they constitute the postsynaptic compartment of the neurons that receives the most excitatory input. The extracellularly shorter variant of the presynaptic cell adhesion molecules neurexins, β-neurexin, has been implicated in various aspects of synaptic function, including neurotransmitter release. However, its role in developing or stabilizing dendritic spines as fundamental computational units of excitatory synapses has remained unclear. Here, we show through morphological analysis that the deletion of β-neurexins in hippocampal neurons in vitro and in hippocampal tissue in vivo affects presynaptic dense-core vesicles, as hypothesized earlier, and, unexpectedly, alters the postsynaptic spine structure. Specifically, we observed that the absence of β-neurexins led to an increase in filopodial-like protrusions in vitro and more mature mushroom-type spines in the CA1 region of adult knockout mice. In addition, the deletion of β-neurexins caused alterations in the spine head dimension and an increase in spines with perforations of their postsynaptic density but no changes in the overall number of spines or synapses. Our results indicate that presynaptic β-neurexins play a role across the synaptic cleft, possibly by aligning with postsynaptic binding partners and glutamate receptors via transsynaptic columns.
Collapse
Affiliation(s)
| | | | - Markus Missler
- Institute of Anatomy and Molecular Neurobiology, University Münster, 48149 Münster, Germany; (L.M.); (J.S.)
| | - Astrid Rohlmann
- Institute of Anatomy and Molecular Neurobiology, University Münster, 48149 Münster, Germany; (L.M.); (J.S.)
| |
Collapse
|
22
|
Ribeiro-Davis A, Al Saeedy DY, Jahr FM, Hawkins E, McClay JL, Deshpande LS. Ketamine Produces Antidepressant Effects by Inhibiting Histone Deacetylases and Upregulating Hippocampal Brain-Derived Neurotrophic Factor Levels in a Diisopropyl Fluorophosphate-Based Rat Model of Gulf War Illness. J Pharmacol Exp Ther 2024; 388:647-654. [PMID: 37863487 PMCID: PMC10801753 DOI: 10.1124/jpet.123.001824] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 09/14/2023] [Accepted: 09/14/2023] [Indexed: 10/22/2023] Open
Abstract
Approximately one-third of Gulf War veterans suffer from Gulf War Illness (GWI), which encompasses mood disorders and depressive symptoms. Deployment-related exposure to organophosphate compounds has been associated with GWI development. Epigenetic modifications have been reported in GWI veterans. We previously showed that epigenetic histone dysregulations were associated with decreased brain-derived neurotrophic factor (BDNF) expression in a GWI rat model. GWI has no effective therapies. Ketamine (KET) has recently been approved by the Food and Drug Administration for therapy-resistant depression. Interestingly, BDNF upregulation underlies KET's antidepressant effect in GWI-related depression. Here, we investigated whether KET's effect on histone mechanisms signals BDNF upregulations in GWI. Male Sprague-Dawley rats were injected once daily with diisopropyl fluorophosphate (DFP; 0.5 mg/kg, s.c., 5 days). At 6 months following DFP exposure, KET (10 mg/kg, i.p.) was injected, and brains were dissected 24 hours later. Western blotting was used for protein expression, and epigenetic studies used chromatin immunoprecipitation methods. Dil staining was conducted for assessing dendritic spines. Our results indicated that an antidepressant dose of KET inhibited the upregulation of histone deacetylase (HDAC) enzymes in DFP rats. Furthermore, KET restored acetylated histone occupancy at the Bdnf promoter IV and induced BDNF protein expression in DFP rats. Finally, KET treatment also increased the spine density and altered the spine diversity with increased T-type and decreased S-type spines in DFP rats. Given these findings, we propose that KET's actions involve the inhibition of HDAC expression, upregulation of BDNF, and dendritic modifications that together ameliorates the pathologic synaptic plasticity and exerts an antidepressant effect in DFP rats. SIGNIFICANCE STATEMENT: This study offers evidence supporting the involvement of epigenetic histone pathways in the antidepressant effects of ketamine (KET) in a rat model of Gulf War Illness (GWI)-like depression. This effect is achieved through the modulation of histone acetylation at the Bdnf promoter, resulting in elevated brain-derived neurotrophic factor expression and subsequent dendritic remodeling in the hippocampus. These findings underscore the rationale for considering KET as a potential candidate for clinical trials aimed at managing GWI-related depression.
Collapse
Affiliation(s)
- Ana Ribeiro-Davis
- Departments of Neurology (A.R.-D., E.H., L.S.D.), Pharmacology and Toxicology (L.S.D.), School of Medicine, Virginia Commonwealth University, Richmond, Virginia and Department of Pharmacotherapy and Outcome Sciences (D.Y.A.S., F.M.J., J.L.M.), School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Dalia Y Al Saeedy
- Departments of Neurology (A.R.-D., E.H., L.S.D.), Pharmacology and Toxicology (L.S.D.), School of Medicine, Virginia Commonwealth University, Richmond, Virginia and Department of Pharmacotherapy and Outcome Sciences (D.Y.A.S., F.M.J., J.L.M.), School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Fay M Jahr
- Departments of Neurology (A.R.-D., E.H., L.S.D.), Pharmacology and Toxicology (L.S.D.), School of Medicine, Virginia Commonwealth University, Richmond, Virginia and Department of Pharmacotherapy and Outcome Sciences (D.Y.A.S., F.M.J., J.L.M.), School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Elisa Hawkins
- Departments of Neurology (A.R.-D., E.H., L.S.D.), Pharmacology and Toxicology (L.S.D.), School of Medicine, Virginia Commonwealth University, Richmond, Virginia and Department of Pharmacotherapy and Outcome Sciences (D.Y.A.S., F.M.J., J.L.M.), School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Joseph L McClay
- Departments of Neurology (A.R.-D., E.H., L.S.D.), Pharmacology and Toxicology (L.S.D.), School of Medicine, Virginia Commonwealth University, Richmond, Virginia and Department of Pharmacotherapy and Outcome Sciences (D.Y.A.S., F.M.J., J.L.M.), School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Laxmikant S Deshpande
- Departments of Neurology (A.R.-D., E.H., L.S.D.), Pharmacology and Toxicology (L.S.D.), School of Medicine, Virginia Commonwealth University, Richmond, Virginia and Department of Pharmacotherapy and Outcome Sciences (D.Y.A.S., F.M.J., J.L.M.), School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
23
|
Jóźwiak-Bębenista M, Sokołowska P, Wiktorowska-Owczarek A, Kowalczyk E, Sienkiewicz M. Ketamine - A New Antidepressant Drug with Anti-Inflammatory Properties. J Pharmacol Exp Ther 2024; 388:134-144. [PMID: 37977808 DOI: 10.1124/jpet.123.001823] [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: 06/30/2023] [Revised: 09/04/2023] [Accepted: 10/04/2023] [Indexed: 11/19/2023] Open
Abstract
Ketamine is a new, potent and rapid-acting antidepressant approved for therapy of treatment-resistant depression, which has a different mechanism of action than currently-available antidepressant therapies. It owes its uniquely potent antidepressant properties to a complex mechanism of action, which currently remains unclear. However, it is thought that it acts by modulating the functioning of the glutamatergic system, which plays an important role in the process of neuroplasticity associated with depression. However, preclinical and clinical studies have also found ketamine to reduce inflammation, either directly or indirectly (by activating neuroprotective branches of the kynurenine pathway), among patients exhibiting higher levels of inflammation. Inflammation and immune system activation are believed to play key roles in the development and course of depression. Therefore, the present work examines the role of the antidepressant effect of ketamine and its anti-inflammatory properties in the treatment of depression. SIGNIFICANCE STATEMENT: The present work examines the relationship between the antidepressant effect of ketamine and its anti-inflammatory properties, and the resulting benefits in treatment-resistant depression (TRD). The antidepressant mechanism of ketamine remains unclear, and there is an urgent need to develop new therapeutic strategies for treatment of depression, particularly TRD.
Collapse
Affiliation(s)
- Marta Jóźwiak-Bębenista
- Department of Pharmacology and Toxicology (M.J.-B., P.S., A.W.-O., E.K.) and Department of Pharmaceutical Microbiology and Microbiological Diagnostics (M.S.), Medical University of Lodz, Lodz, Poland
| | - Paulina Sokołowska
- Department of Pharmacology and Toxicology (M.J.-B., P.S., A.W.-O., E.K.) and Department of Pharmaceutical Microbiology and Microbiological Diagnostics (M.S.), Medical University of Lodz, Lodz, Poland
| | - Anna Wiktorowska-Owczarek
- Department of Pharmacology and Toxicology (M.J.-B., P.S., A.W.-O., E.K.) and Department of Pharmaceutical Microbiology and Microbiological Diagnostics (M.S.), Medical University of Lodz, Lodz, Poland
| | - Edward Kowalczyk
- Department of Pharmacology and Toxicology (M.J.-B., P.S., A.W.-O., E.K.) and Department of Pharmaceutical Microbiology and Microbiological Diagnostics (M.S.), Medical University of Lodz, Lodz, Poland
| | - Monika Sienkiewicz
- Department of Pharmacology and Toxicology (M.J.-B., P.S., A.W.-O., E.K.) and Department of Pharmaceutical Microbiology and Microbiological Diagnostics (M.S.), Medical University of Lodz, Lodz, Poland
| |
Collapse
|
24
|
Koyya P, Manthari RK, Pandrangi SL. Brain-Derived Neurotrophic Factor - The Protective Agent Against Neurological Disorders. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:353-366. [PMID: 37287291 PMCID: PMC11348470 DOI: 10.2174/1871527322666230607110617] [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: 03/28/2023] [Revised: 05/19/2023] [Accepted: 05/29/2023] [Indexed: 06/09/2023]
Abstract
The burden of neurological illnesses on global health is significant. Our perception of the molecular and biological mechanisms underlying intellectual processing and behavior has significantly advanced over the last few decades, laying the groundwork for potential therapies for various neurodegenerative diseases. A growing body of literature reveals that most neurodegenerative diseases could be due to the gradual failure of neurons in the brain's neocortex, hippocampus, and various subcortical areas. Research on various experimental models has uncovered several gene components to understand the pathogenesis of neurodegenerative disorders. One among them is the brain-derived neurotrophic factor (BDNF), which performs several vital functions, enhancing synaptic plasticity and assisting in the emergence of long-term thoughts. The pathophysiology of some neurodegenerative diseases, including Alzheimer's, Parkinson's, Schizophrenia, and Huntington's, has been linked to BDNF. According to numerous research, high levels of BDNF are connected to a lower risk of developing a neurodegenerative disease. As a result, we want to concentrate on BDNF in this article and outline its protective role against neurological disorders.
Collapse
Affiliation(s)
- Prathyusha Koyya
- Department of Biotechnology, GITAM School of Science, Gandhi Institute of Technology and Management (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India
| | - Ram Kumar Manthari
- Department of Biotechnology, GITAM School of Science, Gandhi Institute of Technology and Management (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India
| | - Santhi Latha Pandrangi
- Department of Biochemistry and Bioinformatics, GITAM School of Science, Gandhi Institute of Technology and Management (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India
| |
Collapse
|
25
|
Zagrebelsky M, Korte M. Are TrkB receptor agonists the right tool to fulfill the promises for a therapeutic value of the brain-derived neurotrophic factor? Neural Regen Res 2024; 19:29-34. [PMID: 37488840 PMCID: PMC10479861 DOI: 10.4103/1673-5374.374138] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/20/2023] [Accepted: 03/27/2023] [Indexed: 07/26/2023] Open
Abstract
Brain-derived neurotrophic factor signaling via its receptor tropomyosin receptor kinase B regulates several crucial physiological processes. It has been shown to act in the brain, promoting neuronal survival, growth, and plasticity as well as in the rest of the body where it is involved in regulating for instance aspects of the metabolism. Due to its crucial and very pleiotropic activity, reduction of brain-derived neurotrophic factor levels and alterations in the brain-derived neurotrophic factor/tropomyosin receptor kinase B signaling have been found to be associated with a wide spectrum of neurological diseases. However, because of its poor bioavailability and pharmacological properties, brain-derived neurotrophic factor itself has a very low therapeutic value. Moreover, the concomitant binding of exogenous brain-derived neurotrophic factor to the p75 neurotrophin receptor has the potential to elicit several unwanted and deleterious side effects. Therefore, developing tools and approaches to specifically promote tropomyosin receptor kinase B signaling has become an important goal of translational research. Among the newly developed tools are different categories of tropomyosin receptor kinase B receptor agonist molecules. In this review, we give a comprehensive description of the different tropomyosin receptor kinase B receptor agonist drugs developed so far and of the results of their application in animal models of several neurological diseases. Moreover, we discuss the main benefits of tropomyosin receptor kinase B receptor agonists, concentrating especially on the new tropomyosin receptor kinase B agonist antibodies. The benefits observed both in vitro and in vivo upon application of tropomyosin receptor kinase B receptor agonist drugs seem to predominantly depend on their general neuroprotective activity and their ability to promote neuronal plasticity. Moreover, tropomyosin receptor kinase B agonist antibodies have been shown to specifically bind the tropomyosin receptor kinase B receptor and not p75 neurotrophin receptor. Therefore, while, based on the current knowledge, the tropomyosin receptor kinase B receptor agonists do not seem to have the potential to reverse the disease pathology per se, promoting brain-derived neurotrophic factor/tropomyosin receptor kinase B signaling still has a very high therapeutic relevance.
Collapse
Affiliation(s)
- Marta Zagrebelsky
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Braunschweig, Germany
| | - Martin Korte
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Braunschweig, Germany
- Helmholtz Centre for Infection Research, AG NIND, Braunschweig, Germany
| |
Collapse
|
26
|
Mey M, Bhatta S, Suresh S, Montero Labrador L, Piontkivska H, Casadesus G. The LH receptor regulates hippocampal spatial memory and restores dendritic spine density in ovariectomized APP/PS1 AD mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.22.573087. [PMID: 38187770 PMCID: PMC10769359 DOI: 10.1101/2023.12.22.573087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Activation of the luteinizing hormone receptor (LHCGR) rescues spatial memory function and spine density losses associated with gonadectomy and high circulating gonadotropin levels in females. However, whether this extends to the AD brain or the mechanisms that underlie these benefits remain unknown. To address this question, we delivered the LHCGR agonist human chorionic gonadotropin (hCG) intracerebroventricularly (ICV), under reproductively intact and ovariectomized conditions to mimic the post-menopausal state in the APP/PS1mouse brain. Cognitive function was tested using the Morris water maze task, and hippocampal dendritic spine density, Aβ pathology, and signaling changes associated with these endpoints were determined to address mechanisms. Here we show that central LHCGR activation restored function in ovariectomized APP/PS1 female mice to wild-type levels without altering Aβ pathology. LHCGR activation increased hippocampal dendritic spine density regardless of reproductive status, and this was mediated by BDNF-dependent and independent signaling. We also show that ovariectomy in the APP/PS1 brain elicits an increase in peripherally derived pro-inflammatory genes which are inhibited by LHCGR activation. This may mediate reproductive status specific effects of LHCGR agonism on cognitive function and BDNF expression. Together, this work highlights the relevance of the LHCGR on cognition and its therapeutic potential in the "menopausal" AD brain.
Collapse
|
27
|
Luo Y, Ali T, Liu Z, Gao R, Li A, Yang C, Ling L, He L, Li S. EPO prevents neuroinflammation and relieves depression via JAK/STAT signaling. Life Sci 2023; 333:122102. [PMID: 37769806 DOI: 10.1016/j.lfs.2023.122102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 09/04/2023] [Accepted: 09/15/2023] [Indexed: 10/03/2023]
Abstract
AIMS Erythropoietin (EPO) is a glycoprotein cytokine that exerts therapeutic potential on neurological disorders by promoting neurogenesis and angiogenesis. However, its role as an antidepressant via anti-inflammatory axes is poorly explored. Furthermore, chronic inflammation can induce neuroinflammation, concurrent with depressive-like behaviors that anti-inflammatory and antidepressant agents could avert. Here, we aimed to elucidate the antidepressant potential of Erythropoietin (EPO) in the LPS-induced depression model. MAIN METHODS For in vivo analysis, mice were treated with LPS (2 mg/kg BW), Erythropoietin (EPO) (5000 U/kg/day), (Ruxolitinib,15 mg/kg), and K252a (25 μg/kg). Depressive-like behaviors were confirmed via behavior tests, including OFT, FST, SPT, and TST. Cytokines were measured via ELISA, while IBA-1/GFAP expression was determined by immunofluorescence. Further, the desired gene expression was measured by immunoblotting. For in vitro analysis, BV2 and N2a cell lines were cultured, treated with LPS, EPO, Ruxolitinib, and K252a, collected, and analyzed. KEY FINDINGS LPS treatment significantly induced neuroinflammation accompanied by depression-like behaviors in mice. However, EPO treatment rescued LPS-induced changes by averting cytokine production, secretion, and glial cell activation and reducing depressive-like behaviors in mice. Surprisingly, EPO treatment ameliorated LPS-induced JAK2/STAT5 signaling impairment, as validated by JAK2-antagonism. Furthermore, synaptic and dendritic spine defects and BNDF/TrkB signaling upon LPS administration could be prevented by EPO treatment. SIGNIFICANCE EPO could act as an antidepressant via its anti-inflammatory potential by regulating JAK2/STAT5 signaling.
Collapse
Affiliation(s)
- Yanhua Luo
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China.
| | - Tahir Ali
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China; Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Zizhen Liu
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China.
| | - Ruyan Gao
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China.
| | - Axiang Li
- Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, China.
| | - Canyu Yang
- Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, China.
| | - Li Ling
- Department of Endocrinology, The 6th Affiliated Hospital of Shenzhen University Medical School and Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China.
| | - Liufang He
- Pediatrics Department, Shenzhen University General Hospital, Shenzhen University, Shenzhen, China.
| | - Shupeng Li
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China; Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China; Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
28
|
Gross J, Knipper M, Mazurek B. Candidate Key Proteins in Tinnitus: A Bioinformatic Study of Synaptic Transmission in Spiral Ganglion Neurons. Cell Mol Neurobiol 2023; 43:4189-4207. [PMID: 37736859 PMCID: PMC10661836 DOI: 10.1007/s10571-023-01405-w] [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: 07/29/2023] [Accepted: 08/24/2023] [Indexed: 09/23/2023]
Abstract
To study key proteins associated with changes in synaptic transmission in the spiral ganglion in tinnitus, we build three gene lists from the GeneCard database: 1. Perception of sound (PoS), 2. Acoustic stimulation (AcouStim), and 3. Tinnitus (Tin). Enrichment analysis by the DAVID database resulted in similar Gene Ontology (GO) terms for cellular components in all gene lists, reflecting synaptic structures known to be involved in auditory processing. The STRING protein-protein interaction (PPI) network and the Cytoscape data analyzer were used to identify the top two high-degree proteins (HDPs) and their high-score interaction proteins (HSIPs) identified by the combined score (CS) of the corresponding edges. The top two protein pairs (key proteins) for the PoS are BDNF-GDNF and OTOF-CACNA1D and for the AcouStim process BDNF-NTRK2 and TH-CALB1. The Tin process showed BDNF and NGF as HDPs, with high-score interactions with NTRK1 and NGFR at a comparable level. Compared to the PoS and AcouStim process, the number of HSIPs of key proteins (CS > 90. percentile) increases strongly in Tin. In the PoS and AcouStim networks, BDNF receptor signaling is the dominant pathway, and in the Tin network, the NGF-signaling pathway is of similar importance. Key proteins and their HSIPs are good indicators of biological processes and of signaling pathways characteristic for the normal hearing on the one hand and tinnitus on the other.
Collapse
Affiliation(s)
- Johann Gross
- Tinnitus Center, Charité-Universitätsmedizin Berlin, Berlin, Germany.
- Leibniz Society of Science Berlin, Berlin, Germany.
| | - Marlies Knipper
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
- Leibniz Society of Science Berlin, Berlin, Germany
| | - Birgit Mazurek
- Tinnitus Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
29
|
Mottolese N, Uguagliati B, Tassinari M, Cerchier CB, Loi M, Candini G, Rimondini R, Medici G, Trazzi S, Ciani E. Voluntary Running Improves Behavioral and Structural Abnormalities in a Mouse Model of CDKL5 Deficiency Disorder. Biomolecules 2023; 13:1396. [PMID: 37759796 PMCID: PMC10527551 DOI: 10.3390/biom13091396] [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: 07/18/2023] [Revised: 09/06/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Cyclin-dependent kinase-like 5 (CDKL5) deficiency disorder (CDD) is a rare neurodevelopmental disease caused by mutations in the X-linked CDKL5 gene. CDD is characterized by a broad spectrum of clinical manifestations, including early-onset refractory epileptic seizures, intellectual disability, hypotonia, visual disturbances, and autism-like features. The Cdkl5 knockout (KO) mouse recapitulates several features of CDD, including autistic-like behavior, impaired learning and memory, and motor stereotypies. These behavioral alterations are accompanied by diminished neuronal maturation and survival, reduced dendritic branching and spine maturation, and marked microglia activation. There is currently no cure or effective treatment to ameliorate the symptoms of the disease. Aerobic exercise is known to exert multiple beneficial effects in the brain, not only by increasing neurogenesis, but also by improving motor and cognitive tasks. To date, no studies have analyzed the effect of physical exercise on the phenotype of a CDD mouse model. In view of the positive effects of voluntary running on the brain of mouse models of various human neurodevelopmental disorders, we sought to determine whether voluntary daily running, sustained over a month, could improve brain development and behavioral defects in Cdkl5 KO mice. Our study showed that long-term voluntary running improved the hyperlocomotion and impulsivity behaviors and memory performance of Cdkl5 KO mice. This is correlated with increased hippocampal neurogenesis, neuronal survival, spine maturation, and inhibition of microglia activation. These behavioral and structural improvements were associated with increased BDNF levels. Given the positive effects of BDNF on brain development and function, the present findings support the positive benefits of exercise as an adjuvant therapy for CDD.
Collapse
Affiliation(s)
- Nicola Mottolese
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Beatrice Uguagliati
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Marianna Tassinari
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Camilla Bruna Cerchier
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Manuela Loi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Giulia Candini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Roberto Rimondini
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy
| | - Giorgio Medici
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Stefania Trazzi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Elisabetta Ciani
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
30
|
Solana‐Balaguer J, Campoy‐Campos G, Martín‐Flores N, Pérez‐Sisqués L, Sitjà‐Roqueta L, Kucukerden M, Gámez‐Valero A, Coll‐Manzano A, Martí E, Pérez‐Navarro E, Alberch J, Soriano J, Masana M, Malagelada C. Neuron-derived extracellular vesicles contain synaptic proteins, promote spine formation, activate TrkB-mediated signalling and preserve neuronal complexity. J Extracell Vesicles 2023; 12:e12355. [PMID: 37743539 PMCID: PMC10518375 DOI: 10.1002/jev2.12355] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 07/21/2023] [Indexed: 09/26/2023] Open
Abstract
Extracellular vesicles (EVs) play an important role in intercellular communication as carriers of signalling molecules such as bioactive miRNAs, proteins and lipids. EVs are key players in the functioning of the central nervous system (CNS) by influencing synaptic events and modulating recipient neurons. However, the specific role of neuron-to-neuron communication via EVs is still not well understood. Here, we provide evidence that primary neurons uptake neuron-derived EVs in the soma, dendrites, and even in the dendritic spines, and carry synaptic proteins. Neuron-derived EVs increased spine density and promoted the phosphorylation of Akt and ribosomal protein S6 (RPS6), via TrkB-signalling, without impairing the neuronal network activity. Strikingly, EVs exerted a trophic effect on challenged nutrient-deprived neurons. Altogether, our results place EVs in the spotlight for synaptic plasticity modulation as well as a possible therapeutic tool to fight neurodegeneration.
Collapse
Affiliation(s)
- Julia Solana‐Balaguer
- Departament de Biomedicina, Institut de Neurociències (UBneuro)Universitat de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Genís Campoy‐Campos
- Departament de Biomedicina, Institut de Neurociències (UBneuro)Universitat de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Núria Martín‐Flores
- Departament de Biomedicina, Institut de Neurociències (UBneuro)Universitat de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Leticia Pérez‐Sisqués
- Departament de Biomedicina, Institut de Neurociències (UBneuro)Universitat de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Laia Sitjà‐Roqueta
- Departament de Biomedicina, Institut de Neurociències (UBneuro)Universitat de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Melike Kucukerden
- Departament de Biomedicina, Institut de Neurociències (UBneuro)Universitat de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Ana Gámez‐Valero
- Departament de Biomedicina, Institut de Neurociències (UBneuro)Universitat de BarcelonaBarcelonaSpain
- Biomedical Research Networking Center for Epidemiology and Public Health (CIBERESP)MadridSpain
| | - Albert Coll‐Manzano
- Departament de Biomedicina, Institut de Neurociències (UBneuro)Universitat de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Eulàlia Martí
- Departament de Biomedicina, Institut de Neurociències (UBneuro)Universitat de BarcelonaBarcelonaSpain
- Biomedical Research Networking Center for Epidemiology and Public Health (CIBERESP)MadridSpain
| | - Esther Pérez‐Navarro
- Departament de Biomedicina, Institut de Neurociències (UBneuro)Universitat de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Jordi Alberch
- Departament de Biomedicina, Institut de Neurociències (UBneuro)Universitat de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Jordi Soriano
- Departament de Física de la Matèria CondensadaUniversitat de BarcelonaBarcelonaSpain
- Universitat de Barcelona, Institute of Complex Systems (UBICS)Universitat de BarcelonaBarcelonaSpain
| | - Mercè Masana
- Departament de Biomedicina, Institut de Neurociències (UBneuro)Universitat de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Cristina Malagelada
- Departament de Biomedicina, Institut de Neurociències (UBneuro)Universitat de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| |
Collapse
|
31
|
Jean G, Carton J, Haq K, Musto AE. The role of dendritic spines in epileptogenesis. Front Cell Neurosci 2023; 17:1173694. [PMID: 37601280 PMCID: PMC10433379 DOI: 10.3389/fncel.2023.1173694] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 07/06/2023] [Indexed: 08/22/2023] Open
Abstract
Epilepsy is a chronic central nervous system (CNS) disease associated with high morbidity. To date, there is no known disease-modifying therapy for epilepsy. A leading hypothesis for a mechanism of epileptogenesis is the generation of aberrant neuronal networks. Although the underlying biological mechanism is not clear, scientific evidence indicates that it is associated with a hyperexcitable synchronous neuronal network and active dendritic spine plasticity. Changes in dendritic spine morphology are related to altered expression of synaptic cytoskeletal proteins, inflammatory molecules, neurotrophic factors, and extracellular matrix signaling. However, it remains to be determined if these aberrant dendritic spine formations lead to neuronal hyperexcitability and abnormal synaptic connections or whether they constitute an underlying mechanism of seizure susceptibility. Focusing on dendritic spine machinery as a potential target for medications could limit or reverse the development of epilepsy.
Collapse
Affiliation(s)
- Gary Jean
- Medical Program, School of Medicine, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Joseph Carton
- Medical Program, School of Medicine, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Kaleem Haq
- Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Alberto E. Musto
- Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA, United States
- Department of Neurology, Eastern Virginia Medical School, Norfolk, VA, United States
| |
Collapse
|
32
|
Meftahi GH, Jahromi GP. Biochemical Mechanisms of Beneficial Effects of Beta-Alanine Supplements on Cognition. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:1181-1190. [PMID: 37758316 DOI: 10.1134/s0006297923080114] [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: 03/12/2023] [Revised: 05/20/2023] [Accepted: 05/23/2023] [Indexed: 10/03/2023]
Abstract
Using nutritional interventions to cure and manage psychiatric disorders is a promising tool. In this regard, accumulating documents support strong relationships between the diet and brain health throughout the lifespan. Evidence from animal and human studies demonstrated that β-alanine (Beta-alanine; BA), a natural amino acid, provides several benefits in fight against cognitive decline promoting mental health. This review summarizes and reports state-of-the-art evidence on how BA affects cognitive health and argues existence of potential unrevealed biochemical mechanisms and signaling cascades. There is a growing body of evidence showing that BA supplement has a significant role in mental health mediating increase of the cell carnosine and brain-derived neurotrophic factor (BDNF) content. BDNF is one of the most studied neurotrophins in the mammalian brain, which activates several downstream functional cascades via the tropomyosin-related kinase receptor type B (TrkB). Activation of TrkB induces diverse processes, such as programmed cell death and neuronal viability, dendritic branching growth, dendritic spine formation and stabilization, synaptic development, cognitive-related processes, and synaptic plasticity. Carnosine exerts its main effect via its antioxidant properties. This critical antioxidant also scavenges hypochlorous acid (HOCl), another toxic species produced in mammalian cells. Carnosine regulates transcription of hundreds of genes related to antioxidant mechanisms by increasing expression of the nuclear erythroid 2-related factor 2 (Nrf2) and translocating Nrf2 to the nucleus. Another major protective effect of carnosine on the central nervous system (CNS) is related to its anti-glycating, anti-aggregate activities, anti-inflammatory, metal ion chelator activity, and regulation of pro-inflammatory cytokine secretion. These effects could be associated with the carnosine ability to form complexes with metal ions, particularly with zinc (Zn2+). Thus, it seems that BA via BDNF and carnosine mechanisms may improve brain health and cognitive function over the entire human lifespan.
Collapse
Affiliation(s)
- Gholam Hossein Meftahi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Gila Pirzad Jahromi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
33
|
Kastellakis G, Tasciotti S, Pandi I, Poirazi P. The dendritic engram. Front Behav Neurosci 2023; 17:1212139. [PMID: 37576932 PMCID: PMC10412934 DOI: 10.3389/fnbeh.2023.1212139] [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: 04/25/2023] [Accepted: 07/11/2023] [Indexed: 08/15/2023] Open
Abstract
Accumulating evidence from a wide range of studies, including behavioral, cellular, molecular and computational findings, support a key role of dendrites in the encoding and recall of new memories. Dendrites can integrate synaptic inputs in non-linear ways, provide the substrate for local protein synthesis and facilitate the orchestration of signaling pathways that regulate local synaptic plasticity. These capabilities allow them to act as a second layer of computation within the neuron and serve as the fundamental unit of plasticity. As such, dendrites are integral parts of the memory engram, namely the physical representation of memories in the brain and are increasingly studied during learning tasks. Here, we review experimental and computational studies that support a novel, dendritic view of the memory engram that is centered on non-linear dendritic branches as elementary memory units. We highlight the potential implications of dendritic engrams for the learning and memory field and discuss future research directions.
Collapse
Affiliation(s)
- George Kastellakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Greece
| | - Simone Tasciotti
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Greece
- Department of Biology, University of Crete, Heraklion, Greece
| | - Ioanna Pandi
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Greece
- Department of Biology, University of Crete, Heraklion, Greece
| | - Panayiota Poirazi
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Greece
| |
Collapse
|
34
|
Meng P, Zhang X, Liu TT, Liu J, Luo Y, Xie MX, Yang H, Fang R, Guo DW, Zhong ZY, Wang YH, Ge JW. A whole transcriptome profiling analysis for antidepressant mechanism of Xiaoyaosan mediated synapse loss via BDNF/trkB/PI3K signal axis in CUMS rats. BMC Complement Med Ther 2023; 23:198. [PMID: 37322430 DOI: 10.1186/s12906-023-04000-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 05/15/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Depression is a neuropsychiatric disease resulting from deteriorations of molecular networks and synaptic injury induced by stress. Traditional Chinese formula Xiaoyaosan (XYS) exert antidepressant effect, which was demonstrated by a great many of clinical and basic investigation. However, the exact mechanism of XYS has not yet been fully elucidated. METHODS In this study, chronic unpredictable mild stress (CUMS) rats were used as a model of depression. Behavioral test and HE staining were used to detect the anti-depressant effects of XYS. Furthermore, whole transcriptome sequencing was employed to establish the microRNA (miRNA), long non-coding RNA (lncRNA), circular RNA (circRNA), and mRNA profiles. The biological functions and potential mechanisms of XYS for depression were gathered from the GO and KEGG pathway. Then, constructed the competing endogenous RNA (ceRNA) networks to illustrate the regulatory relationship between non-coding RNA (ncRNA) and mRNA. Additionally, longest dendrite length, total length of dendrites, number of intersections, and density of dendritic spines were detected by Golgi staining. MAP2, PSD-95, SYN were detected by immunofluorescence respectively. BDNF, TrkB, p-TrkB, PI3K, Akt, p-Akt were measured by Western Blotting. RESULTS The results showed that XYS could increase the locomotor activity and sugar preference, decreased swimming immobility time as well as attenuate hippocampal pathological damage. A total of 753 differentially expressed lncRNAs (DElncRNAs), 28 circRNAs (DEcircRNAs), 101 miRNAs (DEmiRNAs), and 477 mRNAs (DEmRNAs) were identified after the treatment of XYS in whole transcriptome sequencing analysis. Enrichment results revealed that XYS could regulate multiple aspects of depression through different synapse or synaptic associated signal, such as neurotrophin signaling and PI3K/Akt signaling pathways. Then, vivo experiments indicated that XYS could promote length, density, intersections of synapses and also increase the expression of MAP2 in hippocampal CA1, CA3 regions. Meanwhile, XYS could increase the expression of PSD-95, SYN in the CA1, CA3 regions of hippocampal by regulating the BDNF/trkB/PI3K signal axis. CONCLUSION The possible mechanism on synapse of XYS in depression was successfully predicted. BDNF/trkB/PI3K signal axis were the potential mechanism of XYS on synapse loss for its antidepressant. Collectively, our results provided novel information about the molecular basis of XYS in treating depression.
Collapse
Affiliation(s)
- Pan Meng
- Hunan University of Chinese Medicine, 300 Xueshi Road, Hanpu Science and Education Park, Yuelu District, Hunan, Changsha, China
| | - Xi Zhang
- The Second People's Hospital of Hunan Province, Changsha, Hunan, China
| | - Tong-Tong Liu
- Hunan University of Chinese Medicine, 300 Xueshi Road, Hanpu Science and Education Park, Yuelu District, Hunan, Changsha, China
| | - Jian Liu
- First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yan Luo
- Hunan University of Chinese Medicine, 300 Xueshi Road, Hanpu Science and Education Park, Yuelu District, Hunan, Changsha, China
| | - Ming-Xia Xie
- Hunan University of Chinese Medicine, 300 Xueshi Road, Hanpu Science and Education Park, Yuelu District, Hunan, Changsha, China
| | - Hui Yang
- First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Rui Fang
- Hunan Academy of Chinese Medicine, Yuelu District, 58 Lushan Road, Changsha, Hunan, China
| | - Dong-Wei Guo
- The Second People's Hospital of Hunan Province, Changsha, Hunan, China
| | - Zi-Yan Zhong
- Hunan University of Chinese Medicine, 300 Xueshi Road, Hanpu Science and Education Park, Yuelu District, Hunan, Changsha, China
| | - Yu-Hong Wang
- Hunan University of Chinese Medicine, 300 Xueshi Road, Hanpu Science and Education Park, Yuelu District, Hunan, Changsha, China.
| | - Jin-Wen Ge
- Hunan Academy of Chinese Medicine, Yuelu District, 58 Lushan Road, Changsha, Hunan, China.
| |
Collapse
|
35
|
Arévalo JC, Deogracias R. Mechanisms Controlling the Expression and Secretion of BDNF. Biomolecules 2023; 13:biom13050789. [PMID: 37238659 DOI: 10.3390/biom13050789] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/19/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Brain-derived nerve factor (BDNF), through TrkB receptor activation, is an important modulator for many different physiological and pathological functions in the nervous system. Among them, BDNF plays a crucial role in the development and correct maintenance of brain circuits and synaptic plasticity as well as in neurodegenerative diseases. The proper functioning of the central nervous system depends on the available BDNF concentrations, which are tightly regulated at transcriptional and translational levels but also by its regulated secretion. In this review we summarize the new advances regarding the molecular players involved in BDNF release. In addition, we will address how changes of their levels or function in these proteins have a great impact in those functions modulated by BDNF under physiological and pathological conditions.
Collapse
Affiliation(s)
- Juan Carlos Arévalo
- Department of Cell Biology and Pathology, Institute of Neurosciences of Castille and Leon (INCyL), University of Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Rubén Deogracias
- Department of Cell Biology and Pathology, Institute of Neurosciences of Castille and Leon (INCyL), University of Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| |
Collapse
|
36
|
Ehinger Y, Soneja D, Phamluong K, Salvi A, Ron D. Identification of Novel BDNF-Specific Corticostriatal Circuitries. eNeuro 2023; 10:ENEURO.0238-21.2023. [PMID: 37156610 PMCID: PMC10198608 DOI: 10.1523/eneuro.0238-21.2023] [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: 05/26/2021] [Revised: 03/23/2023] [Accepted: 04/03/2023] [Indexed: 05/10/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is released from axon terminals originating in the cerebral cortex onto striatal neurons. Here, we characterized BDNF neurons in the corticostriatal circuitry. First, we used BDNF-Cre and Ribotag transgenic mouse lines to label BDNF-positive neurons in the cortex and detected BDNF expression in all the subregions of the prefrontal cortex (PFC). Next, we used a retrograde viral tracing strategy, in combination with BDNF-Cre knock-in mice, to map the cortical outputs of BDNF neurons in the dorsomedial and dorsolateral striatum (DMS and DLS, respectively). We found that BDNF-expressing neurons located in the medial prefrontal cortex (mPFC) project mainly to the DMS, and those located in the primary and secondary motor cortices (M1 and M2, respectively) and agranular insular cortex (AI) project mainly to the DLS. In contrast, BDNF-expressing orbitofrontal cortical (OFC) neurons differentially target the dorsal striatum (DS) depending on their mediolateral and rostrocaudal location. Specifically, the DMS is mainly innervated by the medial and ventral part of the orbitofrontal cortex (MO and VO, respectively), whereas the DLS receives projections specifically from the lateral part of the OFC (LO). Together, our study uncovers previously unknown BDNF corticostriatal circuitries. These findings could have important implications for the role of BDNF signaling in corticostriatal pathways.
Collapse
Affiliation(s)
- Yann Ehinger
- Department of Neurology, University of California, San Francisco, 94143-0663 CA
| | - Drishti Soneja
- Department of Neurology, University of California, San Francisco, 94143-0663 CA
| | - Khanhky Phamluong
- Department of Neurology, University of California, San Francisco, 94143-0663 CA
| | - Alexandra Salvi
- Department of Neurology, University of California, San Francisco, 94143-0663 CA
| | - Dorit Ron
- Department of Neurology, University of California, San Francisco, 94143-0663 CA
| |
Collapse
|
37
|
Manti S, Xerra F, Spoto G, Butera A, Gitto E, Di Rosa G, Nicotera AG. Neurotrophins: Expression of Brain-Lung Axis Development. Int J Mol Sci 2023; 24:ijms24087089. [PMID: 37108250 PMCID: PMC10138985 DOI: 10.3390/ijms24087089] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/08/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Neurotrophins (NTs) are a group of soluble growth factors with analogous structures and functions, identified initially as critical mediators of neuronal survival during development. Recently, the relevance of NTs has been confirmed by emerging clinical data showing that impaired NTs levels and functions are involved in the onset of neurological and pulmonary diseases. The alteration in NTs expression at the central and peripheral nervous system has been linked to neurodevelopmental disorders with an early onset and severe clinical manifestations, often named "synaptopathies" because of structural and functional synaptic plasticity abnormalities. NTs appear to be also involved in the physiology and pathophysiology of several airway diseases, neonatal lung diseases, allergic and inflammatory diseases, lung fibrosis, and even lung cancer. Moreover, they have also been detected in other peripheral tissues, including immune cells, epithelium, smooth muscle, fibroblasts, and vascular endothelium. This review aims to provide a comprehensive description of the NTs as important physiological and pathophysiological players in brain and lung development.
Collapse
Affiliation(s)
- Sara Manti
- Pediatric Unit, Department of Human and Pediatric Pathology "Gaetano Barresi", AOUP G. Martino, University of Messina, Via Consolare Valeria, 1, 98124 Messina, Italy
| | - Federica Xerra
- Pediatric Unit, Department of Human and Pediatric Pathology "Gaetano Barresi", AOUP G. Martino, University of Messina, Via Consolare Valeria, 1, 98124 Messina, Italy
| | - Giulia Spoto
- Unit of Child Neurology and Psychiatry, Department of Human Pathology of the Adult and Developmental Age, "Gaetano Barresi" University of Messina, 98124 Messina, Italy
| | - Ambra Butera
- Unit of Child Neurology and Psychiatry, Department of Human Pathology of the Adult and Developmental Age, "Gaetano Barresi" University of Messina, 98124 Messina, Italy
| | - Eloisa Gitto
- Intensive Pediatric Unit, Department of Human Pathology of the Adult and Developmental Age, "Gaetano Barresi" University of Messina, 98124 Messina, Italy
| | - Gabriella Di Rosa
- Unit of Child Neurology and Psychiatry, Department of Human Pathology of the Adult and Developmental Age, "Gaetano Barresi" University of Messina, 98124 Messina, Italy
| | - Antonio Gennaro Nicotera
- Unit of Child Neurology and Psychiatry, Department of Human Pathology of the Adult and Developmental Age, "Gaetano Barresi" University of Messina, 98124 Messina, Italy
| |
Collapse
|
38
|
Zhuo Y, Fu B, Peng R, Ma C, Xie S, Qiu L. Aptamer-based expansion microscopy platform enables signal-amplified imaging of dendritic spines. Talanta 2023; 260:124541. [PMID: 37087946 DOI: 10.1016/j.talanta.2023.124541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/04/2023] [Accepted: 04/08/2023] [Indexed: 04/25/2023]
Abstract
Super-resolution imaging of dendritic spines (DS) can provide valuable information for mechanistic studies related to synaptic physiology and neural plasticity, but challenged by their small dimension (50-200 nm) below the spatial resolution of conventional optical microscopes. In this work, by combining the molecular recognition specificity of aptamer with high programmability of DNA nanotechnology, we developed an expansion microscopy (ExM) platform for imaging DS with enhanced spatial resolution and amplified signal output. Our results demonstrated that the aptamer probe could specifically bind to DS of primary hippocampal neurons. With physical expansion, the DS structure could be effectively enlarged by 4-5 folds, leading to the generation of more structural information. Meantime, the aptamer binding signal could be readily amplified by the introduction of DNA signal amplification strategy, overcoming the drawback of fluorescence dilution during the ExM treatment. This platform enabled evaluation of ischemia-induced early stroke based on the morphological change of DS, highlighting a promising avenue for studying nanoscale structures in biological systems.
Collapse
Affiliation(s)
- Yuting Zhuo
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Bo Fu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Ruizi Peng
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), The Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Changbei Ma
- School of Life Sciences, Central South University, China
| | - Sitao Xie
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), The Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China.
| | - Liping Qiu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), The Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China.
| |
Collapse
|
39
|
Matosin N, Arloth J, Czamara D, Edmond KZ, Maitra M, Fröhlich AS, Martinelli S, Kaul D, Bartlett R, Curry AR, Gassen NC, Hafner K, Müller NS, Worf K, Rehawi G, Nagy C, Halldorsdottir T, Cruceanu C, Gagliardi M, Gerstner N, Ködel M, Murek V, Ziller MJ, Scarr E, Tao R, Jaffe AE, Arzberger T, Falkai P, Kleinmann JE, Weinberger DR, Mechawar N, Schmitt A, Dean B, Turecki G, Hyde TM, Binder EB. Associations of psychiatric disease and ageing with FKBP5 expression converge on superficial layer neurons of the neocortex. Acta Neuropathol 2023; 145:439-459. [PMID: 36729133 PMCID: PMC10020280 DOI: 10.1007/s00401-023-02541-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 02/03/2023]
Abstract
Identification and characterisation of novel targets for treatment is a priority in the field of psychiatry. FKBP5 is a gene with decades of evidence suggesting its pathogenic role in a subset of psychiatric patients, with potential to be leveraged as a therapeutic target for these individuals. While it is widely reported that FKBP5/FKBP51 mRNA/protein (FKBP5/1) expression is impacted by psychiatric disease state, risk genotype and age, it is not known in which cell types and sub-anatomical areas of the human brain this occurs. This knowledge is critical to propel FKBP5/1-targeted treatment development. Here, we performed an extensive, large-scale postmortem study (n = 1024) of FKBP5/1, examining neocortical areas (BA9, BA11 and ventral BA24/BA24a) derived from subjects that lived with schizophrenia, major depression or bipolar disorder. With an extensive battery of RNA (bulk RNA sequencing, single-nucleus RNA sequencing, microarray, qPCR, RNAscope) and protein (immunoblot, immunohistochemistry) analysis approaches, we thoroughly investigated the effects of disease state, ageing and genotype on cortical FKBP5/1 expression including in a cell type-specific manner. We identified consistently heightened FKBP5/1 levels in psychopathology and with age, but not genotype, with these effects strongest in schizophrenia. Using single-nucleus RNA sequencing (snRNAseq; BA9 and BA11) and targeted histology (BA9, BA24a), we established that these disease and ageing effects on FKBP5/1 expression were most pronounced in excitatory superficial layer neurons of the neocortex, and this effect appeared to be consistent in both the granular and agranular areas examined. We then found that this increase in FKBP5 levels may impact on synaptic plasticity, as FKBP5 gex levels strongly and inversely correlated with dendritic mushroom spine density and brain-derived neurotrophic factor (BDNF) levels in superficial layer neurons in BA11. These findings pinpoint a novel cellular and molecular mechanism that has potential to open a new avenue of FKBP51 drug development to treat cognitive symptoms in psychiatric disorders.
Collapse
Affiliation(s)
- Natalie Matosin
- Department of Translational Research in Psychiatry, Max-Planck Institute of Psychiatry, Munich, Germany.
- Molecular Horizons, School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Northfields Ave, Wollongong, 2522, Australia.
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong, 2522, Australia.
| | - Janine Arloth
- Department of Translational Research in Psychiatry, Max-Planck Institute of Psychiatry, Munich, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Darina Czamara
- Department of Translational Research in Psychiatry, Max-Planck Institute of Psychiatry, Munich, Germany
| | - Katrina Z Edmond
- Molecular Horizons, School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Northfields Ave, Wollongong, 2522, Australia
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong, 2522, Australia
| | - Malosree Maitra
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Anna S Fröhlich
- Department of Translational Research in Psychiatry, Max-Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry, Munich, Germany
| | - Silvia Martinelli
- Department of Translational Research in Psychiatry, Max-Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry, Munich, Germany
| | - Dominic Kaul
- Molecular Horizons, School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Northfields Ave, Wollongong, 2522, Australia
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong, 2522, Australia
| | - Rachael Bartlett
- Molecular Horizons, School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Northfields Ave, Wollongong, 2522, Australia
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong, 2522, Australia
| | - Amber R Curry
- Molecular Horizons, School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Northfields Ave, Wollongong, 2522, Australia
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong, 2522, Australia
| | - Nils C Gassen
- Department of Translational Research in Psychiatry, Max-Planck Institute of Psychiatry, Munich, Germany
- Neurohomeostasis Research Group, Institute of Psychiatry, Clinical Centre, University of Bonn, Bonn, Germany
| | - Kathrin Hafner
- Department of Translational Research in Psychiatry, Max-Planck Institute of Psychiatry, Munich, Germany
| | - Nikola S Müller
- Institute of Computational Biology, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Karolina Worf
- Institute of Computational Biology, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Ghalia Rehawi
- Department of Translational Research in Psychiatry, Max-Planck Institute of Psychiatry, Munich, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Corina Nagy
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | | | - Cristiana Cruceanu
- Department of Translational Research in Psychiatry, Max-Planck Institute of Psychiatry, Munich, Germany
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Miriam Gagliardi
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Nathalie Gerstner
- Department of Translational Research in Psychiatry, Max-Planck Institute of Psychiatry, Munich, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, 85764, Neuherberg, Germany
- International Max Planck Research School for Translational Psychiatry, Munich, Germany
| | - Maik Ködel
- Department of Translational Research in Psychiatry, Max-Planck Institute of Psychiatry, Munich, Germany
| | - Vanessa Murek
- Department of Translational Research in Psychiatry, Max-Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Michael J Ziller
- Department of Translational Research in Psychiatry, Max-Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Elizabeth Scarr
- Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
- Synaptic Neurobiology and Cognition Laboratory, Florey Institute for Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Ran Tao
- The Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Andrew E Jaffe
- The Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Thomas Arzberger
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilians University Munich, Nussbaumstrasse 7, 80336, Munich, Germany
- Centre for Neuropathology and Prion Research, Ludwig-Maximilians University Munich, Nussbaumstrasse 7, 80336, Munich, Germany
| | - Peter Falkai
- Department of Translational Research in Psychiatry, Max-Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilians University Munich, Nussbaumstrasse 7, 80336, Munich, Germany
| | - Joel E Kleinmann
- The Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Daniel R Weinberger
- The Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Andrea Schmitt
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilians University Munich, Nussbaumstrasse 7, 80336, Munich, Germany
- Laboratory of Neuroscience (LIM27), Institute of Psychiatry, University of Sao Paulo, Rua Dr. Ovidio Pires de Campos 785, São Paulo, 05453-010, Brazil
| | - Brian Dean
- Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
- Synaptic Neurobiology and Cognition Laboratory, Florey Institute for Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Thomas M Hyde
- The Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Elisabeth B Binder
- Department of Translational Research in Psychiatry, Max-Planck Institute of Psychiatry, Munich, Germany.
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, USA.
| |
Collapse
|
40
|
Zong W, Lu X, Dong G, Zhang L, Li K. Molecular mechanisms of exercise intervention in alleviating the symptoms of autism spectrum disorder: Targeting the structural alterations of synapse. Front Psychiatry 2023; 14:1096503. [PMID: 37065903 PMCID: PMC10102432 DOI: 10.3389/fpsyt.2023.1096503] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/07/2023] [Indexed: 04/18/2023] Open
Abstract
Autism spectrum disorder (ASD) is a complex and heterogeneous neurodevelopmental disorder characterized by stereotyped behaviors, specific interests, and impaired social and communication skills. Synapses are fundamental structures for transmitting information between neurons. It has been reported that synaptic deficits, such as the increased or decreased density of synapses, may contribute to the onset of ASD, which affects the synaptic function and neuronal circuits. Therefore, targeting the recovery of the synaptic normal structure and function may be a promising therapeutic strategy to alleviate ASD symptoms. Exercise intervention has been shown to regulate the structural plasticity of synapses and improve ASD symptoms, but the underlying molecular mechanisms require further exploration. In this review, we highlight the characteristics of synaptic structural alterations in the context of ASD and the beneficial effects of an exercise intervention on improving ASD symptoms. Finally, we explore the possible molecular mechanisms of improving ASD symptoms through exercise intervention from the perspective of regulating synaptic structural plasticity, which contributes to further optimizing the related strategies of exercise intervention promoting ASD rehabilitation in future.
Collapse
Affiliation(s)
- Wenhao Zong
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
- Department of Sports, Quzhou University, Quzhou, China
- College of Sports and Health, Shandong Sport University, Jinan, China
| | - Xiaowen Lu
- Department of Sports, Quzhou University, Quzhou, China
| | - Guijun Dong
- Department of Sports, Quzhou University, Quzhou, China
| | - Li Zhang
- Guangdong-HongKong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Kefeng Li
- Department of Medicine, Quzhou College of Technology, Quzhou, China
| |
Collapse
|
41
|
Li Y, Guan X, He Y, Jia X, Pan L, Wang Y, Han Y, Zhao R, Yang J, Hou T. ProBDNF signaling is involved in periodontitis-induced depression-like behavior in mouse hippocampus. Int Immunopharmacol 2023; 116:109767. [PMID: 36738676 DOI: 10.1016/j.intimp.2023.109767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/15/2023] [Accepted: 01/19/2023] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Increasing evidence supports the association between periodontitis and depression. However, the specific mechanisms remain to be further elucidated. The present study aimed to mechanistically investigate the regional roles of proBDNF (the precursor of brain-derived neurotrophic factor) in periodontitis induced depression-like behavior in mice. METHODS Experimental periodontitis model was established by periodontal injection of Porphyromonas gingivalis lipopolysaccharide (Pg-LPS) in 8-week-old male Bdnf-HA/HA mice for 3 weeks. The depression-like behaviors, spontaneous exploratory activity and the level of anxiety were assessed by behavior tests. The activation of microglia and astrocytes, as well as the expression of Interleukin (IL)-1β and Tumor necrosis factor (TNF)-α in the hippocampus, prefrontal cortex, and cortex were further assessed by immunofluorescence and western blots. The levels of IL-1β in blood serum and expression of occludin as well as claudin5 in the hippocampus, prefrontal cortex, and cortex were further determined by enzyme-linked immunosorbent assay and western blot. Finally, the expression of proBDNF, its receptors, and mature BDNF (mBDNF), as well as neuronal activity were measured by western blots and immunofluorescence. RESULTS Pg-LPS successfully induced periodontitis in mice and caused obvious depression-like behavior. Furthermore, we observed an increased activation of astrocytes and microglia, as well as a significant increase in expression of IL-1β and TNF-α in the hippocampus of mice treated with Pg-LPS, with elevated level of IL-1β in serum and decreased expression of occludin and claudin5 in the hippocampus. Importantly, we found that the levels of proBDNF and its receptors, SorCS2 and p75NTR, were increased significantly; however, the level of mBDNF was decreased, therefor leading to greater ratio of proBDNF/mBDNF. In addition, we also detected decreased neuronal activity in the hippocampus of mice treated with Pg-LPS. CONCLUSIONS Our results indicate that Pg-LPS-induced periodontitis could cause depression-like behaviors in mice, and the proBDNF signaling is involved in the process.
Collapse
Affiliation(s)
- Yingxue Li
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Xiaoyue Guan
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yani He
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Xiangbin Jia
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Lifei Pan
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yuting Wang
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yue Han
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Rui Zhao
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Jianmin Yang
- Department of Medicine, Weill Cornell Medical School, Cornell University, New York, NY 10065, USA.
| | - Tiezhou Hou
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| |
Collapse
|
42
|
Peregud DI, Baronets VY, Terebilina NN, Gulyaeva NV. Role of BDNF in Neuroplasticity Associated with Alcohol Dependence. BIOCHEMISTRY (MOSCOW) 2023; 88:404-416. [PMID: 37076286 DOI: 10.1134/s0006297923030094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2023]
Abstract
Abstract
Chronic alcohol consumption is characterized by disturbances of neuroplasticity. Brain-derived neurotrophic factor (BDNF) is believed to be critically involved in this process. Here we aimed to review actual experimental and clinical data related to BDNF participation in neuroplasticity in the context of alcohol dependence. As has been shown in experiments with rodents, alcohol consumption is accompanied by the brain region-specific changes of BDNF expression and by structural and behavioral impairments. BDNF reverses aberrant neuroplasticity observed during alcohol intoxication. According to the clinical data parameters associated with BDNF demonstrate close correlation with neuroplastic changes accompanying alcohol dependence. In particular, the rs6265 polymorphism within the BDNF gene is associated with macrostructural changes in the brain, while peripheral BDNF concentration may be associated with anxiety, depression, and cognitive impairment. Thus, BDNF is involved in the mechanisms of alcohol-induced changes of neuroplasticity, and polymorphisms within the BDNF gene and peripheral BDNF concentration may serve as biomarkers, diagnostic or prognostic factors in treatment of alcohol abuse.
Collapse
Affiliation(s)
- Danil I Peregud
- Federal State Budgetary Institution "V. Serbsky National Medical Research Center for Psychiatry and Drug Addiction" of the Ministry of Health of the Russian Federation, Moscow, 119002, Russia.
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia
| | - Valeria Yu Baronets
- Federal State Budgetary Institution "V. Serbsky National Medical Research Center for Psychiatry and Drug Addiction" of the Ministry of Health of the Russian Federation, Moscow, 119002, Russia
| | - Natalia N Terebilina
- Federal State Budgetary Institution "V. Serbsky National Medical Research Center for Psychiatry and Drug Addiction" of the Ministry of Health of the Russian Federation, Moscow, 119002, Russia
| | - Natalia V Gulyaeva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia
- Research and Clinical Center for Neuropsychiatry of Moscow Healthcare Department, Moscow, 115419, Russia
| |
Collapse
|
43
|
Ge X, Zheng M, Hu M, Fang X, Geng D, Liu S, Wang L, Zhang J, Guan L, Zheng P, Xie Y, Pan W, Zhou M, Zhou L, Tang R, Zheng K, Yu Y, Huang XF. Butyrate ameliorates quinolinic acid-induced cognitive decline in obesity models. J Clin Invest 2023; 133:154612. [PMID: 36787221 PMCID: PMC9927952 DOI: 10.1172/jci154612] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/20/2022] [Indexed: 02/15/2023] Open
Abstract
Obesity is a risk factor for neurodegenerative disease associated with cognitive dysfunction, including Alzheimer's disease. Low-grade inflammation is common in obesity, but the mechanism between inflammation and cognitive impairment in obesity is unclear. Accumulative evidence shows that quinolinic acid (QA), a neuroinflammatory neurotoxin, is involved in the pathogenesis of neurodegenerative processes. We investigated the role of QA in obesity-induced cognitive impairment and the beneficial effect of butyrate in counteracting impairments of cognition, neural morphology, and signaling. We show that in human obesity, there was a negative relationship between serum QA levels and cognitive function and decreased cortical gray matter. Diet-induced obese mice had increased QA levels in the cortex associated with cognitive impairment. At single-cell resolution, we confirmed that QA impaired neurons, altered the dendritic spine's intracellular signal, and reduced brain-derived neurotrophic factor (BDNF) levels. Using Caenorhabditis elegans models, QA induced dopaminergic and glutamatergic neuron lesions. Importantly, the gut microbiota metabolite butyrate was able to counteract those alterations, including cognitive impairment, neuronal spine loss, and BDNF reduction in both in vivo and in vitro studies. Finally, we show that butyrate prevented QA-induced BDNF reductions by epigenetic enhancement of H3K18ac at BDNF promoters. These findings suggest that increased QA is associated with cognitive decline in obesity and that butyrate alleviates neurodegeneration.
Collapse
Affiliation(s)
- Xing Ge
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Mingxuan Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Minmin Hu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Xiaoli Fang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Deqin Geng
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Sha Liu
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Li Wang
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Jun Zhang
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Li Guan
- The Second Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Peng Zheng
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medical, Indigenous, and Health, University of Wollongong, New South Wales, Australia
| | - Yuanyi Xie
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medical, Indigenous, and Health, University of Wollongong, New South Wales, Australia
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Menglu Zhou
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Limian Zhou
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Renxian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Xu-Feng Huang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China.,Illawarra Health and Medical Research Institute (IHMRI) and School of Medical, Indigenous, and Health, University of Wollongong, New South Wales, Australia
| |
Collapse
|
44
|
Powers RM, Hevner RF, Halpain S. The Neuron Navigators: Structure, function, and evolutionary history. Front Mol Neurosci 2023; 15:1099554. [PMID: 36710926 PMCID: PMC9877351 DOI: 10.3389/fnmol.2022.1099554] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 12/19/2022] [Indexed: 01/13/2023] Open
Abstract
Neuron navigators (Navigators) are cytoskeletal-associated proteins important for neuron migration, neurite growth, and axon guidance, but they also function more widely in other tissues. Recent studies have revealed novel cellular functions of Navigators such as macropinocytosis, and have implicated Navigators in human disorders of axon growth. Navigators are present in most or all bilaterian animals: vertebrates have three Navigators (NAV1-3), Drosophila has one (Sickie), and Caenorhabditis elegans has one (Unc-53). Structurally, Navigators have conserved N- and C-terminal regions each containing specific domains. The N-terminal region contains a calponin homology (CH) domain and one or more SxIP motifs, thought to interact with the actin cytoskeleton and mediate localization to microtubule plus-end binding proteins, respectively. The C-terminal region contains two coiled-coil domains, followed by a AAA+ family nucleoside triphosphatase domain of unknown activity. The Navigators appear to have evolved by fusion of N- and C-terminal region homologs present in simpler organisms. Overall, Navigators participate in the cytoskeletal response to extracellular cues via microtubules and actin filaments, in conjunction with membrane trafficking. We propose that uptake of fluid-phase cues and nutrients and/or downregulation of cell surface receptors could represent general mechanisms that explain Navigator functions. Future studies developing new models, such as conditional knockout mice or human cerebral organoids may reveal new insights into Navigator function. Importantly, further biochemical studies are needed to define the activities of the Navigator AAA+ domain, and to study potential interactions among different Navigators and their binding partners.
Collapse
Affiliation(s)
- Regina M. Powers
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, United States,Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| | - Robert F. Hevner
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States,Department of Pathology, UC San Diego School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Shelley Halpain
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, United States,Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States,*Correspondence: Shelley Halpain, ✉
| |
Collapse
|
45
|
von Bohlen Und Halbach O. Neurotrophic Factors and Dendritic Spines. ADVANCES IN NEUROBIOLOGY 2023; 34:223-254. [PMID: 37962797 DOI: 10.1007/978-3-031-36159-3_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] [Indexed: 11/15/2023]
Abstract
Dendritic spines are highly dynamic structures that play important roles in neuronal plasticity. The morphologies and the numbers of dendritic spines are highly variable, and this diversity is correlated with the different morphological and physiological features of this neuronal compartment. Dendritic spines can change their morphology and number rapidly, allowing them to adapt to plastic changes. Neurotrophic factors play important roles in the brain during development. However, these factors are also necessary for a variety of processes in the postnatal brain. Neurotrophic factors, especially members of the neurotrophin family and the ephrin family, are involved in the modulation of long-lasting effects induced by neuronal plasticity by acting on dendritic spines, either directly or indirectly. Thereby, the neurotrophic factors play important roles in processes attributed, for example, to learning and memory.
Collapse
|
46
|
18β-Glycyrrhetinic Acid Ameliorates Neuroinflammation Linked Depressive Behavior Instigated by Chronic Unpredictable Mild Stress via Triggering BDNF/TrkB Signaling Pathway in Rats. Neurochem Res 2023; 48:551-569. [PMID: 36307572 PMCID: PMC9616426 DOI: 10.1007/s11064-022-03779-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/06/2022] [Accepted: 09/30/2022] [Indexed: 02/04/2023]
Abstract
Evidence shows that inflammatory responses may encompass the onset of severe depressive illness. Traditionally used licorice contains 18β-glycyrrhetinic acid (18βGA), which has been demonstrated to reduce inflammation and oxidative stress. This study investigates the antidepressant effects of 18βGA and the underlying mechanism in rats exposed to chronic unpredictable mild stress (CUMS). Wistar rats were exposed to CUMS for 36 consecutive days to establish depression. 18βGA (10, 20, and 50 mg/kg) or fluoxetine was given once daily (from day 30 to day 36). Thereafter, behavior parameters (sucrose preference test, forced-swimming test, open-field test, body weight), pro-inflammatory cytokines, neurotransmitters, adrenocorticotropic hormone (ACTH), corticosterone (CORT), and liver biomarkers were studied. Immunohistochemistry and western blot analyses were conducted to investigate the protein's expression. 18βGA (20 and 50 mg/kg) treatment increased sucrose intake, locomotion in the open-field test, decreased immobility time in the forced swim test, and improved body weight in CUMS-exposed rats. The therapy of 18βGA dramatically declined cytokines, ACTH and CORT and improved 5HT and norepinephrine in CUMS rats. Furthermore, BDNF and TrkB proteins were down-regulated in CUMS group, which was increased to varying degrees by 18βGA at doses of 20 and 50 mg/kg. Therefore, 18βGA ameliorates depressive-like behavior persuaded by chronic unpredictable mild stress, decreases neuroinflammation, liver biomarkers, stress hormones, and improves body weight, brain neurotransmitter concentration via activating on BDNF/TrkB signaling pathway in both PFC and hippocampus in rats.
Collapse
|
47
|
Russo L, Giacomelli C, Fortino M, Marzo T, Ferri G, Calvello M, Viegi A, Magrì A, Pratesi A, Pietropaolo A, Cardarelli F, Martini C, Rizzarelli E, Marchetti L, La Mendola D, Trincavelli ML. Neurotrophic Activity and Its Modulation by Zinc Ion of a Dimeric Peptide Mimicking the Brain-Derived Neurotrophic Factor N-Terminal Region. ACS Chem Neurosci 2022; 13:3453-3463. [PMID: 36346920 PMCID: PMC9732821 DOI: 10.1021/acschemneuro.2c00463] [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] [Indexed: 11/09/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a neurotrophin (NT) essential for neuronal development and synaptic plasticity. Dysregulation of BDNF signaling is implicated in different neurological disorders. The direct NT administration as therapeutics has revealed to be challenging. This has prompted the design of peptides mimicking different regions of the BDNF structure. Although loops 2 and 4 have been thoroughly investigated, less is known regarding the BDNF N-terminal region, which is involved in the selective recognition of the TrkB receptor. Herein, a dimeric form of the linear peptide encompassing the 1-12 residues of the BDNF N-terminal (d-bdnf) was synthesized. It demonstrated to act as an agonist promoting specific phosphorylation of TrkB and downstream ERK and AKT effectors. The ability to promote TrkB dimerization was investigated by advanced fluorescence microscopy and molecular dynamics (MD) simulations, finding activation modes shared with BDNF. Furthermore, d-bdnf was able to sustain neurite outgrowth and increase the expression of differentiation (NEFM, LAMC1) and polarization markers (MAP2, MAPT) demonstrating its neurotrophic activity. As TrkB activity is affected by zinc ions in the synaptic cleft, we first verified the ability of d-bdnf to coordinate zinc and then the effect of such complexation on its activity. The d-bdnf neurotrophic activity was reduced by zinc complexation, demonstrating the role of the latter in tuning the activity of the new peptido-mimetic. Taken together our data uncover the neurotrophic properties of a novel BDNF mimetic peptide and pave the way for future studies to understand the pharmacological basis of d-bdnf action and develop novel BDNF-based therapeutic strategies.
Collapse
Affiliation(s)
- Lara Russo
- Dipartimento
di Farmacia, Università di Pisa, Pisa 56127, Italy
| | | | | | - Tiziano Marzo
- Dipartimento
di Farmacia, Università di Pisa, Pisa 56127, Italy
| | - Gianmarco Ferri
- Laboratorio
NEST, Scuola Normale Superiore, Pisa 56127, Italy
| | | | | | - Antonio Magrì
- Istituto
di Cristallografia, Consiglio Nazionale delle Ricerche (CNR), Catania 95126, Italy
| | - Alessandro Pratesi
- Dipartimento
di Chimica e Chimica Industriale, Università
di Pisa, Pisa 56124, Italy
| | | | | | - Claudia Martini
- Dipartimento
di Farmacia, Università di Pisa, Pisa 56127, Italy
| | - Enrico Rizzarelli
- Istituto
di Cristallografia, Consiglio Nazionale delle Ricerche (CNR), Catania 95126, Italy,Università
degli Studi di Catania, Catania 95124, Italy
| | - Laura Marchetti
- Dipartimento
di Farmacia, Università di Pisa, Pisa 56127, Italy,
| | - Diego La Mendola
- Dipartimento
di Farmacia, Università di Pisa, Pisa 56127, Italy,
| | | |
Collapse
|
48
|
Alfonsetti M, d’Angelo M, Castelli V. Neurotrophic factor-based pharmacological approaches in neurological disorders. Neural Regen Res 2022; 18:1220-1228. [PMID: 36453397 PMCID: PMC9838155 DOI: 10.4103/1673-5374.358619] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Aging is a physiological event dependent on multiple pathways that are linked to lifespan and processes leading to cognitive decline. This process represents the major risk factor for aging-related diseases such as Alzheimer's disease, Parkinson's disease, and ischemic stroke. The incidence of all these pathologies increases exponentially with age. Research on aging biology has currently focused on elucidating molecular mechanisms leading to the development of those pathologies. Cognitive deficit and neurodegeneration, common features of aging-related pathologies, are related to the alteration of the activity and levels of neurotrophic factors, such as brain-derived neurotrophic factor, nerve growth factor, and glial cell-derived neurotrophic factor. For this reason, treatments that modulate neurotrophin levels have acquired a great deal of interest in preventing neurodegeneration and promoting neural regeneration in several neurological diseases. Those treatments include both the direct administration of neurotrophic factors and the induced expression with viral vectors, neurotrophins' binding with biomaterials or other molecules to increase their bioavailability but also cell-based therapies. Considering neurotrophins' crucial role in aging pathologies, here we discuss the involvement of several neurotrophic factors in the most common brain aging-related diseases and the most recent therapeutic approaches that provide direct and sustained neurotrophic support.
Collapse
Affiliation(s)
- Margherita Alfonsetti
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Michele d’Angelo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy,Correspondence to: Vanessa Castelli, .
| |
Collapse
|
49
|
Habit Formation and the Effect of Repeated Stress Exposures on Cognitive Flexibility Learning in Horses. Animals (Basel) 2022; 12:ani12202818. [DOI: 10.3390/ani12202818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/13/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
Horse training exposes horses to an array of cognitive and ethological challenges. Horses are routinely required to perform behaviours that are not aligned to aspects of their ethology, which may delay learning. While horses readily form habits during training, not all of these responses are considered desirable, resulting in the horse being subject to retraining. This is a form of cognitive flexibility and is critical to the extinction of habits and the learning of new responses. It is underpinned by complex neural processes which can be impaired by chronic or repeated stress. Domestic horses may be repeatedly exposed to multiples stressors. The potential contribution of stress impairments of cognitive flexibility to apparent training failures is not well understood, however research from neuroscience can be used to understand horses’ responses to training. We trained horses to acquire habit-like responses in one of two industry-style aversive instrumental learning scenarios (moving away from the stimulus-instinctual or moving towards the stimulus-non-instinctual) and evaluated the effect of repeated stress exposures on their cognitive flexibility in a reversal task. We measured heart rate as a proxy for noradrenaline release, salivary cortisol and serum Brain Derived Neurotrophic Factor (BDNF) to infer possible neural correlates of the learning outcomes. The instinctual task which aligned with innate equine escape responses to aversive stimuli was acquired significantly faster than the non-instinctual task during both learning phases, however contrary to expectations, the repeated stress exposure did not impair the reversal learning. We report a preliminary finding that serum BDNF and salivary cortisol concentrations in horses are positively correlated. The ethological salience of training tasks and cognitive flexibility learning can significantly affect learning in horses and trainers should adapt their practices where such tasks challenge innate equine behaviour.
Collapse
|
50
|
Tassinari M, Mottolese N, Galvani G, Ferrara D, Gennaccaro L, Loi M, Medici G, Candini G, Rimondini R, Ciani E, Trazzi S. Luteolin Treatment Ameliorates Brain Development and Behavioral Performance in a Mouse Model of CDKL5 Deficiency Disorder. Int J Mol Sci 2022; 23:ijms23158719. [PMID: 35955854 PMCID: PMC9369425 DOI: 10.3390/ijms23158719] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/28/2022] [Accepted: 08/03/2022] [Indexed: 12/16/2022] Open
Abstract
CDKL5 deficiency disorder (CDD), a rare and severe neurodevelopmental disease caused by mutations in the X-linked CDKL5 gene, is characterized by early-onset epilepsy, intellectual disability, and autistic features. Although pharmacotherapy has shown promise in the CDD mouse model, safe and effective clinical treatments are still far off. Recently, we found increased microglial activation in the brain of a mouse model of CDD, the Cdkl5 KO mouse, suggesting that a neuroinflammatory state, known to be involved in brain maturation and neuronal dysfunctions, may contribute to the pathophysiology of CDD. The present study aims to evaluate the possible beneficial effect of treatment with luteolin, a natural flavonoid known to have anti-inflammatory and neuroprotective activities, on brain development and behavior in a heterozygous Cdkl5 (+/−) female mouse, the mouse model of CDD that best resembles the genetic clinical condition. We found that inhibition of neuroinflammation by chronic luteolin treatment ameliorates motor stereotypies, hyperactive profile and memory ability in Cdkl5 +/− mice. Luteolin treatment also increases hippocampal neurogenesis and improves dendritic spine maturation and dendritic arborization of hippocampal and cortical neurons. These findings show that microglia overactivation exerts a harmful action in the Cdkl5 +/− brain, suggesting that treatments aimed at counteracting the neuroinflammatory process should be considered as a promising adjuvant therapy for CDD.
Collapse
Affiliation(s)
- Marianna Tassinari
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Nicola Mottolese
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Giuseppe Galvani
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Domenico Ferrara
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Laura Gennaccaro
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Manuela Loi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Giorgio Medici
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Giulia Candini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Roberto Rimondini
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy
| | - Elisabetta Ciani
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Stefania Trazzi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| |
Collapse
|