1
|
Wei XY, Xu FP, Chen XB, Xu Q, Deng LY, Sun C, Chen XY, Tan F, Yang ZM. Peiyuan Ningshen-ointment alleviates learning and memory impairment induced by sleep deprivation through activating the cAMP/PKA signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156469. [PMID: 40315642 DOI: 10.1016/j.phymed.2025.156469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/19/2024] [Accepted: 02/03/2025] [Indexed: 05/04/2025]
Abstract
BACKGROUND Sleep Deprivation (SD) seriously impacts the brain's learning and memory functioning, and the treatment methods and their efficacy are limited. PYNS ointment (Peiyuan Ningshen-ointment) is an herbal ointment consisting of eleven Chinese herbs, which have been proven to have positive therapeutic effects on learning and memory impairment. However, its mechanism of action is still unclear. PURPOSE The purpose of this research was to explore the efficacy and the underlying mechanism of PYNS ointment in learning and memory impairment caused by SD. METHODS PYNS ointment chemical composition was analyzed using UHPLC-Q Exactive HRMS. The SD model was established by intraperitoneal injection of DL-4-chlorophenylalanine. The effects of PYNS ointment were verified by behavioral experiments and histological observation. Furthermore, the potential underlying mechanisms of action of PYNS ointment were explored using immunological techniques, RNA-Seq, and molecular biology experiments. RESULT PYNS ointment protected against SD-induced learning and memory impairment by reducing hippocampal neuronal damage, regulating synaptic plasticity, and inhibiting the inflammatory response. Mechanismly, PYNS ointment alleviated astrocyte reactive hyperplasia and morphological damage. Furthermore, PYNS ointment promoted the expression and polarization of aquaporin-4 (AQP4) through activation of the cAMP/PKA signaling pathway. While enhancing Aβ clearance, it also acted on the downstream of CREB/BDNF to regulate PSD95 and increase synaptic plasticity. CONCLUSION PYNS ointment significantly improved hippocampal damage and enhanced learning and memory function in SD rats by inhibiting inflammation, reducing astrocyte reactivity, and enhancing synaptic plasticity through upregulation of the cAMP/PKA/AQP4/BDNF pathway. Our findings provide new insights into the mechanisms underlying learning and memory decline caused by sleep deprivation.
Collapse
Affiliation(s)
- Xiao-Yu Wei
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China
| | - Fu-Ping Xu
- Famous Traditional Chinese Medicine Inheritance Studio of Guangdong Province, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China
| | - Xiang-Bin Chen
- The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510720, PR China
| | - Qing Xu
- The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510720, PR China
| | - Lu-Yao Deng
- The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510720, PR China
| | - Chen Sun
- Famous Traditional Chinese Medicine Inheritance Studio of Guangdong Province, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China
| | - Xin-Yan Chen
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine 510120, Guangzhou, PR China
| | - Fei Tan
- Famous Traditional Chinese Medicine Inheritance Studio of Guangdong Province, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China
| | - Zhi-Min Yang
- Famous Traditional Chinese Medicine Inheritance Studio of Guangdong Province, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China.
| |
Collapse
|
2
|
Khan S, Khan MM, Badruddeen, Ahmad U, Akhtar W, Islam A. Exploring NMDAR pathways in ischemic stroke: implications for neurotoxic and neuroprotective mechanisms and therapeutic strategies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04357-8. [PMID: 40490524 DOI: 10.1007/s00210-025-04357-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2025] [Accepted: 06/02/2025] [Indexed: 06/11/2025]
Abstract
Stroke is one of the leading causes of disability and mortality worldwide, with ischemic stroke representing the most prevalent and devastating form. This review offers an in-depth exploration of the critical role of N-Methyl-D-Aspartate Receptor (NMDAR) signaling in mediating the brain's response to ischemic injury. NMDAR activation triggers glutamate excitotoxicity, setting off a cascade of neurotoxic events that lead to mitochondrial dysfunction and the generation of reactive oxygen species (ROS). These damaging processes not only intensify neuronal injury but also activate apoptotic pathways, including p53-mediated and Notch signaling. Furthermore, the review highlights necroptosis as a key cell death mechanism in ischemic injury and examines the subsequent disruption of the blood-brain barrier (BBB), which exacerbates brain damage. In the context of neuroprotective signaling, we explore the distinct roles of synaptic and extrasynaptic NMDAR activation, neurotrophic factor-mediated signaling, and the intricate crosstalk between neurotoxic and neuroprotective pathways. This review also explores novel hypotheses and emerging perspectives in NMDAR-mediated ischemic stroke, highlighting potential mechanisms and therapeutic implications. Additionally, it covers cutting-edge experimental approaches to investigate NMDAR function in stroke and provides critical insights into conflicting findings in NMDAR research, addressing key controversies and their impact on future studies. Therapeutic strategies targeting ischemic stroke are critically examined, with an emphasis on potential interventions that could mitigate the effects of ischemia. The review also highlights ongoing clinical trials investigating novel therapeutic approaches and outlines the future direction of ischemic stroke therapy. This comprehensive review offers a deep understanding of the complex molecular mechanisms involved in ischemic stroke via NMDAR and provides valuable insights into the promising therapeutic avenues that could lead to more effective treatments.
Collapse
Affiliation(s)
- Sara Khan
- Department of Pharmacy, Integral University, Kursi Road, Dasauli, Lucknow, 226026, Uttar Pradesh, India
| | - Mohd Muazzam Khan
- Department of Pharmacy, Integral University, Kursi Road, Dasauli, Lucknow, 226026, Uttar Pradesh, India.
| | - Badruddeen
- Department of Pharmacy, Integral University, Kursi Road, Dasauli, Lucknow, 226026, Uttar Pradesh, India
| | - Usama Ahmad
- Department of Pharmacy, Integral University, Kursi Road, Dasauli, Lucknow, 226026, Uttar Pradesh, India
| | - Wasim Akhtar
- Department of Pharmacy, Integral University, Kursi Road, Dasauli, Lucknow, 226026, Uttar Pradesh, India
| | - Anas Islam
- Department of Pharmacy, Integral University, Kursi Road, Dasauli, Lucknow, 226026, Uttar Pradesh, India
| |
Collapse
|
3
|
Liu Y, Takamatsu Y, Chen K, Ding Y, Oka Y, Sugiyama T, Maejima H. Skilled reaching training combined with pharmacological inhibition of histone deacetylases potentiated motor recovery after intracerebral hemorrhage in a synergic manner. Brain Res 2025; 1856:149569. [PMID: 40081517 DOI: 10.1016/j.brainres.2025.149569] [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: 12/11/2024] [Revised: 02/19/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Neuronal recovery after stroke is supported by the expression of genes involved in neuronal plasticity and neuroprotection. As an epigenetic modification, histone acetylation modulates gene expression elicited by neurorehabilitation. This study aimed to investigate the combined effects of skilled reaching training (SRT) and the pharmacological inhibition of histone deacetylase (HDAC) using sodium butyrate (NaB) on motor function recovery after intracerebral hemorrhage (ICH). Wistar rats were divided into five groups: Sham, ICH, ICH plus SRT (ICH + SRT), ICH plus NaB administration (ICH + NaB), and ICH plus SRT plus NaB administration (ICH + SRT + NaB). ICH surgery was conducted based on the microinjection of collagenase into the striatum near the internal capsule. NaB treatment (300 mg/kg injected intraperitoneally) and SRT were performed five days a week for four weeks after ICH surgery, followed by tissue collection. After the intervention, the ICH + SRT + NaB group exhibited significant improvement in skilled motor function, accompanied by a significant increase in neurotrophin 4 and synaptophysin expression in the ipsilateral motor cortex. This study showed that combination therapy of SRT and HDAC inhibition synergistically promoted motor recovery after ICH, accompanied by the upregulation of crucial genes for neuroplasticity. Taken together, this study indicates that HDAC inhibition could represent an enriched neuronal platform for neurorehabilitation after ICH.
Collapse
Affiliation(s)
- Yushan Liu
- Graduate School of Health Sciences, Hokkaido University, Kita 12 Nishi 5, Kita-ku, Sapporo 060-0812, Japan
| | - Yasuyuki Takamatsu
- Department of Rehabilitation Science, Faculty of Health Sciences, Hokkaido University, Kita 12 Nishi 5, Kita-ku, Sapporo 060-0812, Japan; Department of Physical Therapy, College of Life and Health Sciences, Chubu University, 1200 Matsumoto-cho, Kasugai 487-8501, Japan
| | - Ke Chen
- Graduate School of Health Sciences, Hokkaido University, Kita 12 Nishi 5, Kita-ku, Sapporo 060-0812, Japan
| | - Yuan Ding
- Graduate School of Health Sciences, Hokkaido University, Kita 12 Nishi 5, Kita-ku, Sapporo 060-0812, Japan
| | - Yuichiro Oka
- Department of Rehabilitation Science, Faculty of Health Sciences, Hokkaido University, Kita 12 Nishi 5, Kita-ku, Sapporo 060-0812, Japan
| | - Takuya Sugiyama
- Graduate School of Health Sciences, Hokkaido University, Kita 12 Nishi 5, Kita-ku, Sapporo 060-0812, Japan
| | - Hiroshi Maejima
- Department of Rehabilitation Science, Faculty of Health Sciences, Hokkaido University, Kita 12 Nishi 5, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
4
|
Mitrovic M, Selakovic D, Jovicic N, Ljujic B, Rosic G. BDNF/proBDNF Interplay in the Mediation of Neuronal Apoptotic Mechanisms in Neurodegenerative Diseases. Int J Mol Sci 2025; 26:4926. [PMID: 40430064 PMCID: PMC12112594 DOI: 10.3390/ijms26104926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2025] [Revised: 05/12/2025] [Accepted: 05/20/2025] [Indexed: 05/29/2025] Open
Abstract
The neurotrophic system includes neurotrophins, such as brain-derived neurotrophic factor (BDNF) and its precursor proBDNF, which play conflicting roles in neuronal survival and apoptosis, with their balance having a significant impact on neurodegenerative outcomes. While BDNF is widely acknowledged as a potent neurotrophin that promotes neuronal survival and differentiation, its precursor, proBDNF, has the opposite effect, promoting apoptosis and neuronal death. This review highlights the new and unique aspects of BDNF/proBDNF interaction in the modulation of neuronal apoptotic pathways in neurodegenerative disorders. It systematically discusses the cross-talk in apoptotic signaling at the molecular level, whereby BDNF activates survival pathways such as PI3K/Akt and MAPK/ERK, whereas proBDNF activates p75NTR and sortilin to induce neuronal apoptosis via JNK, RhoA, NFkB, and Rac-GTPase pathways such as caspase activation and mitochondrial injury. Moreover, this review emphasizes the factors that affect the balance between proBDNF and BDNF levels within the context of neurodegeneration, including proteolytic processing, the expression of TrkB and p75NTR receptors, and extrinsic gene transcription regulators. Cellular injury, stress, or signaling pathway alterations can disrupt the balance of BDNF/proBDNF, which may be involved in apoptotic-related neurodegenerative diseases like Alzheimer's, Parkinson's, and Huntington's diseases. This review provides a comprehensive framework for targeting neurotrophin signaling in the development of innovative therapies for neuronal survival and managing apoptotic-related neurodegenerative disorders, addressing the mechanistic complexity and clinical feasibility of BDNF/proBDNF interaction.
Collapse
Affiliation(s)
- Marina Mitrovic
- Department of Medical Biochemistry, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Dragica Selakovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Nemanja Jovicic
- Department of Histology and Embryology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Biljana Ljujic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Gvozden Rosic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| |
Collapse
|
5
|
Qiu Y, Zhu L, Cai W, Zhu L. Research Progress on BDNF and Depression. ACS Chem Neurosci 2025. [PMID: 40359301 DOI: 10.1021/acschemneuro.5c00193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025] Open
Abstract
Depression is a potentially life-threatening psychiatric disorder that affects the physical and mental health of millions of individuals worldwide. It can manifest at any stage of life, inducing profound emotional despondency, negative cognitions, and, in severe cases, suicidal ideation, often accompanied by physical symptoms, bringing a significant burden on both families and society. Brain-derived neurotrophic factor (BDNF), a member of the neurotrophic factor family, is widely expressed in the central nervous system (CNS), particularly in regions, such as the hippocampus and cortex. Numerous studies have shown that an imbalance or inadequate conversion of pro-brain-derived neurotrophic factor (proBDNF) into its mature form, mature BDNF (mBDNF), may impair neuronal plasticity, which is crucial to the pathogenesis of depression. This paper provides a comprehensive review of the neurotrophic mechanisms implicated in depression, covering the location, expression, and release of BDNF; the relationship between proBDNF, mBDNF, and depression; and the downstream signaling pathways triggered by BNDF binding to its receptors. This review aims to provide a theoretical foundation for understanding the pathogenesis and clinical treatment of depression.
Collapse
Affiliation(s)
- Yahong Qiu
- The Key Laboratory of Developmental Genes and Human Disease, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Lixia Zhu
- Patent Examination Cooperation (JIANGSU) Center of the Patent Office, China National Intellectual Property Administration (CNIPA), Suzhou, Jiangsu 215163, China
| | - Wenyan Cai
- The Key Laboratory of Developmental Genes and Human Disease, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Lijuan Zhu
- The Key Laboratory of Developmental Genes and Human Disease, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| |
Collapse
|
6
|
Al-Ashram MM, Nader MA, El-Sheakh AR. Role of sacubitril/valsartan in modulating diabetes mediated cognitive and neuronal impairment. Int Immunopharmacol 2025; 154:114431. [PMID: 40157081 DOI: 10.1016/j.intimp.2025.114431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 02/19/2025] [Accepted: 03/03/2025] [Indexed: 04/01/2025]
Abstract
Earlier investigations had established that Diabetes mellitus (DM) caused significant damage in the central nervous system, bringing about diabetic encephalopathy and increasing the risk of cognitive-related problems. Nonetheless, the inherent pathophysiology of cognitive dysfunctions in DM is not well understood. The current study aimed to examine the possible influences of sacubitril/valsartan (SAC/VAL), an angiotensin receptor blocker/neprilysin inhibitor (ARNI), on cognitive dysfunction associated with streptozotocin (STZ)-induced diabetic rats. SAC/VAL and VAL treatments were initiated three days after the diabetic condition was established and continued daily for eight weeks. Normal, non-diabetic rats were reserved as a control group. Both SAC/VAL and VAL treatment in diabetic rats ameliorated diabetes induced oxidative stress as indicated by reduced malondialdehyde (MDA), increased total antioxidant capacity (TAO) in hippocampal tissue and decreased serum advanced glycation end products (AGEs), also inflammatory and apoptotic changes were observed and proved by the reduction of tumor necrosis factor alpha (TNF-α) and caspase -3 in rat hippocampus. SAC/VAL administration to diabetic rats also improved neuronal damages as reflected by restored cAMP response element-binding protein (CREB), brain derived neurotrophic factor (BDNF) and pre-synaptic phosphoproteins, synapsin I and growth associated protein-43 (GAP-43) in the hippocampus of diabetic rats. Additionally, SAC/VAL treated diabetic rats markedly reduced signs of cognitive deterioration during the Morris water maze test. Collectively, these findings suggested that SAC/VAL might play a vital role in improvement of the cognitive impairment observed in diabetic rats through antioxidant, anti-inflammatory and anti-apoptotic actions.
Collapse
Affiliation(s)
- Mai M Al-Ashram
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansura University, Mansura, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansura National University, Gamasa, Egypt.
| | - Manar A Nader
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansura University, Mansura, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansura National University, Gamasa, Egypt
| | - Ahmed R El-Sheakh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansura University, Mansura, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansura National University, Gamasa, Egypt; Future studies and Risks management, National Committee of Drugs, Academy of Scientific Research, Ministry of Higher Education, Elsayeda Zeinab, Egypt
| |
Collapse
|
7
|
Wang J, Schupp W, Sakata K. Peripheral tissue BDNF expression is affected by promoter IV defect and enriched environments in mice: negative hippocampus-intestine and positive thymus-serum-muscle correlations. Mol Med 2025; 31:164. [PMID: 40316902 PMCID: PMC12048937 DOI: 10.1186/s10020-025-01196-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 04/02/2025] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) expression is reduced in the brain of various central nervous system (CNS) disorders, but its relation to peripheral expression remains unclear. This study aimed to determine peripheral BDNF expression affected by BDNF promoter IV defect and enriched environment treatment (EET). Promoter IV defect is associated with CNS disorders and chronic stress, whereas EET increases hippocampal BDNF expression and ameliorates CNS dysfunctions. METHODS Enzyme-linked immunosorbent assay measured BDNF protein levels in eleven regions (hippocampus, frontal cortex, heart, lung, liver, spleen, intestine, kidney, intestine, thymus, muscle, serum) in wild-type and knock-in promoter IV (KIV) mice with or without 3 weeks of EET provided after weaning. RESULTS Knock-in promoter IV resulted in BDNF levels significantly decreased in muscle, but significantly increased in intestine, liver, thymus, and serum, which suggests compensatory upregulation of other promoters in those tissues. EET increased BDNF levels in muscle and serum of KIV mice and thymus of wild-type mice, suggesting EET's beneficial effects in muscle motor and adaptive immune regulation. EET increased hippocampal BDNF levels in both genotypes, which significantly negatively correlated with intestine BDNF levels, suggesting its role in the brain-gut axis. EET reduced wild-type heart BDNF levels, possibly through parasympathetic regulation. Significant positive BDNF correlations were observed among serum-muscle, serum-thymus, lung-spleen, and intestine-liver, suggesting inter-organ interaction and regulation of BDNF. Partial Least Squares discriminant analyses (PLS-DA) identified that variations in BDNF levels in intestine, liver, frontal cortex, and serum contribute most to classify promoter IV defect, and those in hippocampus, serum, heart, thymus, and liver contribute most to classify EET effects. CONCLUSION This is the first study to demonstrate how genetic and environmental factors affect BDNF expression in peripheral tissues, highlighting the complex BDNF correlations across organ systems and suggesting usefulness of multivariate BDNF analyses for detecting promoter IV defect and enriched environment effects. Elucidation of BDNF's role and regulatory mechanisms in peripheral organ systems may help better our understanding of its connection to CNS disorders and their treatments.
Collapse
Affiliation(s)
- Janet Wang
- Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, 71 S. Manassas St. Room 225N, Memphis, TN, 38103, USA
- Department of Psychiatry, College of Medicine, University of Tennessee Health Science Center, Memphis, USA
| | - William Schupp
- Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, 71 S. Manassas St. Room 225N, Memphis, TN, 38103, USA
| | - Kazuko Sakata
- Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, 71 S. Manassas St. Room 225N, Memphis, TN, 38103, USA.
- Department of Psychiatry, College of Medicine, University of Tennessee Health Science Center, Memphis, USA.
| |
Collapse
|
8
|
Raviv T, Shteinfer-Kuzmine A, Moyal MM, Shoshan-Barmatz V. Resveratrol's Pro-Apoptotic Effects in Cancer Are Mediated Through the Interaction and Oligomerization of the Mitochondrial VDAC1. Int J Mol Sci 2025; 26:3963. [PMID: 40362204 PMCID: PMC12071565 DOI: 10.3390/ijms26093963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Revised: 04/10/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
Resveratrol is a naturally occurring phenolic compound found in various foods such as red wine, chocolate, peanuts, and blueberries. Both in-vitro and in-vivo studies have shown that it has a broad spectrum of pharmacological effects such as providing cellular protection and promoting longevity. These effects include antioxidant, anti-inflammatory, neuroprotective, and anti-viral properties, as well as improvements in cardio-metabolic health and anti-aging benefits. Additionally, resveratrol has demonstrated the ability to induce cell death and inhibit tumor growth across different types and stages of cancer. However, the dual effects of resveratrol-acting to support cell survival in some contexts, while inducing cell death in others-is still not fully understood. In this study, we identify a novel target for resveratrol: the voltage-dependent anion channel 1 (VDAC1), a multi-functional outer mitochondrial membrane protein that plays a key role in regulating both cell survival and death. Our findings show that resveratrol increased VDAC1 expression levels and promoted its oligomerization, leading to apoptotic cell death. Additionally, resveratrol elevated intracellular Ca2+ levels and enhanced the production of reactive oxygen species (ROS). Resveratrol also induced the detachment of hexokinase I from VDAC1, a key enzyme in metabolism, and regulating apoptosis. When VDAC1 expression was silenced using specific siRNA, resveratrol-induced cell death was significantly reduced, indicating that VDAC1 is essential for its pro-apoptotic effects. Additionally, both resveratrol and its analog, trans-2,3,5,4'-tetrahydroxystilbene-2-O-glucoside (TSG), directly interacted with purified VDAC1, as revealed by microscale thermophoresis, with similar binding affinities. However, unlike resveratrol, TSG did not induce VDAC1 overexpression or apoptosis. These results demonstrate that resveratrol-induced apoptosis is linked to increased VDAC1 expression and its oligomerization. This positions resveratrol not only as a protective agent, but also as a pro-apoptotic compound. Consequently, resveratrol offers a promising therapeutic approach for cancer, with potentially fewer side effects compared to conventional treatments, due to its natural origins in plants and food products.
Collapse
Affiliation(s)
- Tal Raviv
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel;
| | - Anna Shteinfer-Kuzmine
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (A.S.-K.); (M.M.M.)
| | - Meital M. Moyal
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (A.S.-K.); (M.M.M.)
| | - Varda Shoshan-Barmatz
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel;
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (A.S.-K.); (M.M.M.)
| |
Collapse
|
9
|
Carter AC, Koreman GT, Petrocelli JE, Robb JE, Bushinsky EM, Trowbridge SK, Kingsley DM, Walsh CA, Song JHT, Greenberg ME. FOS binding sites are a hub for the evolution of activity-dependent gene regulatory programs in human neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.646366. [PMID: 40236085 PMCID: PMC11996375 DOI: 10.1101/2025.03.31.646366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
After birth, sensory inputs to neurons trigger the induction of activity-dependent genes (ADGs) that mediate many aspects of neuronal maturation and plasticity. To identify human-specific ADGs, we characterized these genes in human-chimpanzee tetraploid neurons. We identified 235 ADGs that are differentially expressed between human and chimpanzee neurons and found that their nearby regulatory sites are species-biased in their binding of the transcription factor FOS. An assessment of these sites revealed that many are enriched for single nucleotide variants that promote or eliminate FOS binding in human neurons. Disrupting the function of individual species-biased FOS-bound enhancers diminishes expression of nearby genes and affects the firing dynamics of human neurons. Our findings indicate that FOS-bound enhancers are frequent sites of evolution and that they regulate human-specific ADGs that may contribute to the unusually protracted and complex process of postnatal human brain development.
Collapse
|
10
|
Michael A, Onisiforou A, Georgiou P, Koumas M, Powels C, Mammadov E, Georgiou AN, Zanos P. (2R,6R)-hydroxynorketamine prevents opioid abstinence-related negative affect and stress-induced reinstatement in mice. Br J Pharmacol 2025. [PMID: 40155780 DOI: 10.1111/bph.70018] [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: 11/22/2024] [Revised: 01/09/2025] [Accepted: 02/05/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND AND PURPOSE Opioid use disorder (OUD) is a pressing public health concern marked by frequent relapse during periods of abstinence, perpetuated by negative affective states. Classical antidepressants or the currently prescribed opioid pharmacotherapies have limited efficacy to reverse the negative affect or prevent relapse. EXPERIMENTAL APPROACH Using mouse models, we investigated the effects of ketamine's metabolite (2R,6R)-hydroxynorketamine (HNK) on reversing conditioning to sub-effective doses of morphine in stress-susceptible mice, preventing conditioned-place aversion and alleviating acute somatic abstinence symptoms in opioid-dependent mice. Additionally, we evaluated its effects on anhedonia, anxiety-like behaviours and cognitive impairment during protracted opioid abstinence, while mechanistic studies examined cortical EEG oscillations and synaptic plasticity markers. KEY RESULTS (2R,6R)-HNK reversed conditioning to sub-effective doses of morphine in stress-susceptible mice and prevented conditioned-place aversion and acute somatic abstinence symptoms in opioid-dependent mice. In addition, (2R,6R)-HNK reversed anhedonia, anxiety-like behaviours and cognitive impairment emerging during protracted opioid abstinence plausibly via a restoration of impaired cortical high-frequency EEG oscillations, through a GluN2A-NMDA receptor-dependent mechanism. Notably, (2R,6R)-HNK facilitated the extinction of opioid conditioning, prevented stress-induced reinstatement of opioid-seeking behaviours and reduced the propensity for enhanced morphine self-consumption in mice previously exposed to opioids. CONCLUSIONS AND IMPLICATIONS These findings emphasize the therapeutic potential of (2R,6R)-HNK, which is currently in Phase II clinical trials, in addressing stress-related opioid responses. Reducing the time and cost required for development of new medications for the treatment of OUDs via drug repurposing is critical due to the opioid crisis we currently face.
Collapse
Affiliation(s)
- Andria Michael
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
- Center for Applied Neuroscience (CAN), University of Cyprus, Nicosia, Cyprus
| | - Anna Onisiforou
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
- Center for Applied Neuroscience (CAN), University of Cyprus, Nicosia, Cyprus
| | - Polymnia Georgiou
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, USA
| | - Morfeas Koumas
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| | - Chris Powels
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Elmar Mammadov
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Andrea N Georgiou
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
- Center for Applied Neuroscience (CAN), University of Cyprus, Nicosia, Cyprus
| | - Panos Zanos
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
- Center for Applied Neuroscience (CAN), University of Cyprus, Nicosia, Cyprus
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| |
Collapse
|
11
|
Singh AA, Katiyar S, Song M. Phytochemicals Targeting BDNF Signaling for Treating Neurological Disorders. Brain Sci 2025; 15:252. [PMID: 40149774 PMCID: PMC11939912 DOI: 10.3390/brainsci15030252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025] Open
Abstract
Neurological disorders are defined by a deterioration or disruption of the nervous system's structure and function. These diseases, which include multiple sclerosis, Alzheimer's disease, Parkinson's disease, Huntington's disease, and schizophrenia, are caused by intricate pathological processes that include excitotoxicity, neuroinflammation, oxidative stress, genetic mutations, and compromised neurotrophic signaling. Although current pharmaceutical treatments relieve symptoms, their long-term efficacy is limited due to adverse side effects and weak neuroprotective properties. However, when combined with other neuroprotective drugs or adjunct therapy, they may offer additional benefits and improve treatment outcomes. Phytochemicals have emerged as attractive therapeutic agents due to their ability to regulate essential neurotrophic pathways, especially the brain-derived neurotrophic factor (BDNF) signaling cascade. BDNF is an important target for neurodegenerative disease (ND) treatment since it regulates neuronal survival, synaptic plasticity, neurogenesis, and neuroprotection. This review emphasizes the molecular pathways through which various phytochemicals-such as flavonoids, terpenoids, alkaloids, and phenolic compounds-stimulate BDNF expression and modulate its downstream signaling pathways, including GSK-3β, MAPK/ERK, PI3K/Akt/mTOR, CREB, and Wnt/β-catenin. This paper also highlights how phytochemical combinations may interact to enhance BDNF activity, offering new therapeutic options for ND treatment. Despite their potential for neuroprotection, phytochemicals face challenges related to pharmacokinetics, blood-brain barrier (BBB) permeability, and absorption, highlighting the need for further research into combination therapies and improved formulations. Clinical assessment and mechanistic understanding of BDNF-targeted phytotherapy should be the main goals of future studies. The therapeutic efficacy of natural compounds in regulating neurotrophic signaling is highlighted in this review, providing a viable approach to the prevention and treatment of NDs.
Collapse
Affiliation(s)
- Alka Ashok Singh
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | - Shweta Katiyar
- Department of Botany, SBN Government PG College, Barwani 451551, MP, India;
| | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| |
Collapse
|
12
|
Zhang D, He X, Wang Y, Wang X, Han X, Liu H, Xing Y, Jiang B, Xiu Z, Bao Y, Dong Y. Hesperetin-Enhanced Metformin to Alleviate Cognitive Impairment via Gut-Brain Axis in Type 2 Diabetes Rats. Int J Mol Sci 2025; 26:1923. [PMID: 40076550 PMCID: PMC11900253 DOI: 10.3390/ijms26051923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Diabetes constitutes a risk factor for cognitive impairment, whereas insulin resistance serves as the shared pathogenesis underlying both diabetes and cognitive decline. The use of metformin for treating cognitive impairment remains controversial. The present study found that hesperetin, a flavanone derived from citrus peel, enhanced metformin's efficacy in reducing blood sugar levels, improving insulin sensitivity, and ameliorating cognitive impairment in diabetic rats. Additionally, it reduced the required dosage of metformin to one-third of its conventional dose. Transcriptome analysis and 16S rRNA sequencing revealed that the activation of insulin and cyclic-adenosine monophosphate response element binding protein (CREB)/brain-derived neurotrophic factor (BDNF) pathways benefited from the regulation of gut microbiota and the promotion of short-chain fatty acid (SCFA) producers such as Romboutsia. Furthermore, this study demonstrated that hesperetin supplementation counteracted the upregulation of β-site amyloid precursor protein cleaving enzyme 1 (BACE1), a pathological factor of Alzheimer's disease (AD) that was induced by metformin. Our findings reveal that hesperetin can be used in supplementary treatment for cognitive impairment associated with diabetes.
Collapse
Affiliation(s)
- Danyang Zhang
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (D.Z.); (X.H.); (X.W.); (X.H.); (H.L.); (Y.X.); (B.J.); (Z.X.); (Y.B.)
| | - Xiaoshi He
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (D.Z.); (X.H.); (X.W.); (X.H.); (H.L.); (Y.X.); (B.J.); (Z.X.); (Y.B.)
| | - Yinbo Wang
- Dianxi Research Institute, Dalian University of Technology, Baoshan 678000, China;
| | - Xiaoyu Wang
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (D.Z.); (X.H.); (X.W.); (X.H.); (H.L.); (Y.X.); (B.J.); (Z.X.); (Y.B.)
| | - Xiao Han
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (D.Z.); (X.H.); (X.W.); (X.H.); (H.L.); (Y.X.); (B.J.); (Z.X.); (Y.B.)
| | - Haodong Liu
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (D.Z.); (X.H.); (X.W.); (X.H.); (H.L.); (Y.X.); (B.J.); (Z.X.); (Y.B.)
| | - Yan Xing
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (D.Z.); (X.H.); (X.W.); (X.H.); (H.L.); (Y.X.); (B.J.); (Z.X.); (Y.B.)
| | - Bo Jiang
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (D.Z.); (X.H.); (X.W.); (X.H.); (H.L.); (Y.X.); (B.J.); (Z.X.); (Y.B.)
| | - Zhilong Xiu
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (D.Z.); (X.H.); (X.W.); (X.H.); (H.L.); (Y.X.); (B.J.); (Z.X.); (Y.B.)
| | - Yongming Bao
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (D.Z.); (X.H.); (X.W.); (X.H.); (H.L.); (Y.X.); (B.J.); (Z.X.); (Y.B.)
- School of Ocean Science and Technology, Dalian University of Technology, Panjin 124221, China
| | - Yuesheng Dong
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (D.Z.); (X.H.); (X.W.); (X.H.); (H.L.); (Y.X.); (B.J.); (Z.X.); (Y.B.)
| |
Collapse
|
13
|
K Soman S, Swain M, Dagda RK. BDNF-TrkB Signaling in Mitochondria: Implications for Neurodegenerative Diseases. Mol Neurobiol 2025; 62:1756-1769. [PMID: 39030441 PMCID: PMC11909598 DOI: 10.1007/s12035-024-04357-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 07/09/2024] [Indexed: 07/21/2024]
Abstract
Brain-derived neurotrophic factor (BDNF) plays a pivotal role in neuronal development, synaptic plasticity, and overall neuronal health by binding to its receptor, tyrosine receptor kinase B (TrkB). This review delves into the intricate mechanisms through which BDNF-TrkB signaling influences mitochondrial function and potentially influences pathology in neurodegenerative diseases. This review highlights the BDNF-TrkB signaling pathway which regulates mitochondrial bioenergetics, biogenesis, and dynamics, mitochondrial processes vital for synaptic transmission and plasticity. Furthermore, we explore how the BDNF-TrkB-PKA signaling in the cytosol and in mitochondria affects mitochondrial transport and distribution and mitochondrial content, which is crucial for supporting the energy demands of synapses. The dysregulation of this signaling pathway is linked to various neurodegenerative diseases, including Alzheimer's and Parkinson's disease, which are characterized by mitochondrial dysfunction and reduced BDNF expression. By examining seminal studies that have characterized this signaling pathway in health and disease, the present review underscores the potential of enhancing BDNF-TrkB signaling to mitigate mitochondrial dysfunction in neurodegenerative diseases, offering insights into therapeutic strategies to enhance neuronal resilience and function.
Collapse
Affiliation(s)
- Smijin K Soman
- Department of Pharmacology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA
| | - Maryann Swain
- Department of Pharmacology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA
| | - Ruben K Dagda
- Department of Pharmacology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA.
| |
Collapse
|
14
|
Yin D, Zhao L, Deng S, Xie Y, Ro KS, Yang Z, Du L, Xie J, Wei D. Lactiplantibacillus plantarum X7022 Plays Roles on Aging Mice with Memory Impairment Induced by D-Galactose Through Restoring Neuronal Damage, Relieving Inflammation and Oxidative Stress. Probiotics Antimicrob Proteins 2025; 17:1-14. [PMID: 38183568 DOI: 10.1007/s12602-023-10208-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2023] [Indexed: 01/08/2024]
Abstract
In this study, Lactiplantibacillus plantarum X7022 was applied to ameliorate memory impairment of aging mice induced by D-galactose. The strain showed specific choloylglycine hydrolysis ability based on in vitro investigation. Morris water maze test showed L. plantarum X7022 administration improved learning ability and spatial memory of aging mice. The gavage of L. plantarum X7022 displayed a promising ability of relieving cerebral oxidative stress and hippocampal inflammatory condition according to the increased GSH level and SOD activity and decreased MDA level, as well as decreased TNF-α, IL-1β, and IL-6 levels. The intervention with the strain could protect neuron by regulating cell apoptosis and AChE overexpression and inhibiting amyloid-β deposition, as well as affect neuron functions by regulating CREB-BDNF signaling pathways and iNOS expression. Besides, the strain could improve fecal SCFA contents and increase the abundance of anti-inflammatory and antioxidant-related genera such as Lactobacillus, Akkermansia, and Adlercreutzia. These results suggest that L. plantarum X7022 could be a prospective therapeutic alternative for the improvement of memory impairment among the elderly.
Collapse
Affiliation(s)
- Deyi Yin
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China
| | - Li Zhao
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China
| | - Sijing Deng
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China
| | - Yaqi Xie
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China
| | - Kum-Song Ro
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China
- Department of Biotechnology, Faculty of Life Science, Kim Hyong Jik University of Education, Pyongyang, 999093, Democratic People's Republic of Korea
| | - Zeyong Yang
- Department of Anesthesiology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Embryo Original Disease, Shanghai Municipal Key Clinical Specialty, Shanghai, 200030, People's Republic of China
| | - Lei Du
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China.
| | - Jingli Xie
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China.
- Shanghai Collaborative Innovation Center for Biomanufacturing (SCICB), Shanghai, 200237, People's Republic of China.
| | - Dongzhi Wei
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Technology, School of Biotechnology, East China University of Science and Technology, 130 # Meilong Rd, Shanghai, 200237, People's Republic of China
- Shanghai Collaborative Innovation Center for Biomanufacturing (SCICB), Shanghai, 200237, People's Republic of China
| |
Collapse
|
15
|
Zent KH, Dell'Acqua ML. Synapse-to-Nucleus ERK→CREB Transcriptional Signaling Requires Dendrite-to-Soma Ca 2+ Propagation Mediated by L-Type Voltage-Gated Ca 2+ Channels. J Neurosci 2025; 45:e1216242024. [PMID: 39562039 PMCID: PMC11756630 DOI: 10.1523/jneurosci.1216-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/02/2024] [Accepted: 11/09/2024] [Indexed: 11/21/2024] Open
Abstract
The cAMP-response element-binding protein (CREB) transcription factor controls the expression of the neuronal immediate early genes c-fos, Arc, and Bdnf and is essential for long-lasting synaptic plasticity underlying learning and memory. Despite this critical role, there is still ongoing debate regarding the synaptic excitation-transcription (E-T) coupling mechanisms mediating CREB activation in the nucleus. Here we employed optical uncaging of glutamate to mimic synaptic excitation of distal dendrites in conjunction with simultaneous imaging of intracellular Ca2+ dynamics and transcriptional reporter gene expression to elucidate CREB E-T coupling mechanisms in hippocampal neurons cultured from both male and female rats. Using this approach, we found that CREB-dependent transcription was engaged following dendritic stimulation of N-methyl-d-aspartate receptors (NMDARs) only when Ca2+ signals propagated to the soma via subsequent activation of L-type voltage-gated Ca2+ channels resulting in activation of extracellular signal-regulated kinase MAP kinase signaling to sustain CREB phosphorylation in the nucleus. In contrast, dendrite-restricted Ca2+ signals generated by NMDARs failed to stimulate CREB-dependent transcription. Furthermore, Ca2+-CaM-dependent kinase-mediated signaling pathways that may transiently contribute to CREB phosphorylation following stimulation were ultimately dispensable for downstream CREB-dependent transcription and c-Fos induction. These findings emphasize the essential role that L-type Ca2+ channels play in rapidly relaying signals over long distances from synapses located on distal dendrites to the nucleus to control gene expression.
Collapse
Affiliation(s)
- Katlin H Zent
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
- Neuroscience Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Mark L Dell'Acqua
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
- Neuroscience Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
- Neurotechnology Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| |
Collapse
|
16
|
Shibata T, Ihara D, Kirihara Y, Yagi T, Tabuchi A, Kuroda S. Expression of c-fos in cortical neuron cultures under dynamic magnetic field is not suppressed by calcium channel blockers. Drug Discov Ther 2025; 18:391-396. [PMID: 39662933 DOI: 10.5582/ddt.2024.01077] [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: 12/13/2024]
Abstract
Previously, we developed a dynamic magnetic field (DMF) device using neodymium magnets that induced c-fos expression in cortical neurons, while activity-regulated cytoskeleton-associated protein (Arc), and brain-derived neurotrophic factor (BDNF) remained unaffected. The precise signal transduction pathway for c-fos induction under DMF was unclear. This study aimed to investigate the mechanism of immediate early gene (IEG) induction using calcium channel blockers (CCBs). Six experiments were conducted with cortical neurons, employing an NMDA receptor antagonist and an L-type voltage-dependent calcium channel blocker as CCBs. Neuronal cultures were exposed to DMF, CCBs, or both, and IEG expression (Arc, c-fos, BDNF) was measured through polymerase chain reaction. Results showed a tendency for increased c-fos expression with DMF exposure, which was unaffected by CCBs. In contrast, Arc and BDNF were not induced under DMF exposure but were significantly inhibited by CCBs. These findings suggest that c-fos induction under DMF involves a distinct pathway, potentially relevant to stress resistance and drug discovery.
Collapse
Affiliation(s)
- Takashi Shibata
- Department of Neurosurgery, University of Toyama, Japan
- Department of Neurosurgery, Toyama Nishi General Hospital, Japan
| | - Daisuke Ihara
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Yuji Kirihara
- Department of Mechanical Engineering, Institute of Science Tokyo, Tokyo, Japan
| | - Tohru Yagi
- Department of Mechanical Engineering, Institute of Science Tokyo, Tokyo, Japan
| | - Akiko Tabuchi
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | | |
Collapse
|
17
|
Fan Y, Luan X, Wang X, Li H, Zhao H, Li S, Li X, Qiu Z. Exploring the association between BDNF related signaling pathways and depression: A literature review. Brain Res Bull 2025; 220:111143. [PMID: 39608613 DOI: 10.1016/j.brainresbull.2024.111143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/13/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024]
Abstract
Depression is a debilitating mental disease that inflicts significant harm upon individuals and society, yet effective treatment options remain elusive. At present, the pathogenesis of multiple depression is not fully clear, but its occurrence can be related to biological or environmental pathways, among which Brain-derived neurotrophic factor (BDNF) can unequivocally act on two downstream receptors, tyrosine kinase receptor (TrkB) and the p75 neurotrophin receptor (p75NTR), then affect the related signal pathways, affecting the occurrence and development of depression. Accumulating studies have revealed that BDNF-related pathways are critical in the pathophysiology of depression, and their interaction can further influence the efficacy of depression treatment. In this review, we mainly summarized the signaling pathways associated with BDNF and classified them according to different receptors and related molecules, providing promising insights and future directions in the treatment of depression.
Collapse
Affiliation(s)
- Yuchen Fan
- Interventional Medical Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China; Qingdao medical college, Qingdao University, Qingdao, Shandong, China.
| | - Xinchi Luan
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| | - Xuezhe Wang
- Qingdao medical college, Qingdao University, Qingdao, Shandong, China.
| | - Hongchi Li
- Qingdao medical college, Qingdao University, Qingdao, Shandong, China.
| | - Hongjiao Zhao
- Qingdao medical college, Qingdao University, Qingdao, Shandong, China.
| | - Sheng Li
- Qingdao medical college, Qingdao University, Qingdao, Shandong, China.
| | - Xiaoxuan Li
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| | - Zhenkang Qiu
- Interventional Medical Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
18
|
Xu S, Chen J, Xu C, Xu Y, Xu L, Zhao M, Xu T, Cao Y, Li P, Han Z. 2-BFI protects against ischemic stroke by selectively acting on NR2B-containing NMDA receptors. Brain Res 2024; 1845:149284. [PMID: 39423961 DOI: 10.1016/j.brainres.2024.149284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/06/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND AND PURPOSE The intricate roles of NMDA receptors, specifically those containing the NR2A or NR2B subunit, in ischemic stroke pathology necessitate targeted therapeutic investigations. Building on our prior discovery showcasing the neuroprotective potential of 2-(benzofuran-2-yl)-2-imidazoline (2-BFI), an imidazoline I2 receptor ligand, in inhibiting NMDA receptor currents during ischemic stroke, this study aims to elucidate the specific impact of 2-BFI on NR2A- and NR2B-containing NMDARs. EXPERIMENTAL APPROACH Through whole-cell patch-clamp techniques, we observed an inhibition by 2-BFI on NR2A-containing NMDAR currents (IC50 = 238.6 μM) and NR2B-containing NMDAR currents (IC50 = 18.47 μM). Experiments with HEK293 cells expressing exogenous receptor subunits revealed a significantly higher affinity of 2-BFI towards NR2B-containing NMDARs. In vivo studies involved the co-administration of 2-BFI and the NR2A subunit antagonist NVP-AAM077 in rats subjected to transient middle cerebral artery occlusion (tMCAO). Key results 2-BFI exhibited a pronounced preference for inhibiting NR2B-containing NMDAR currents, leading to a notable mitigation of cerebral ischemic injury when administered in conjunction with NVP-AAM077 in the tMCAO rat model. Furthermore, alterations in the expression of downstream proteins specific to NR2B-containing NMDA receptors were observed, suggesting targeted molecular effects. Conclusion and implications This study unveils the neuroprotective potential of 2-BFI in ischemic stroke by selectively inhibiting NR2B-containing NMDA receptors. These findings lay the foundation for precise therapeutic strategies, showcasing the differential roles of NR2A and NR2B subunits and paving the way for advancements in targeted interventions for ischemic stroke treatment.
Collapse
Affiliation(s)
- Shasha Xu
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China; Department of Critical Care Medicine, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Jiaou Chen
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chunfei Xu
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ye Xu
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lu Xu
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Meiqi Zhao
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tong Xu
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yungang Cao
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Peijun Li
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhao Han
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
19
|
Belousova E, Salikhova D, Maksimov Y, Nebogatikov V, Sudina A, Goldshtein D, Ustyugov A. Proposed Mechanisms of Cell Therapy for Alzheimer's Disease. Int J Mol Sci 2024; 25:12378. [PMID: 39596443 PMCID: PMC11595163 DOI: 10.3390/ijms252212378] [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: 10/17/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's disease is a progressive neurodegenerative disorder characterized by mitochondria dysfunction, accumulation of beta-amyloid plaques, and hyperphosphorylated tau tangles in the brain leading to memory loss and cognitive deficits. There is currently no cure for this condition, but the potential of stem cells for the therapy of neurodegenerative pathologies is actively being researched. This review discusses preclinical and clinical studies that have used mouse models and human patients to investigate the use of novel types of stem cell treatment approaches. The findings provide valuable insights into the applications of stem cell-based therapies and include the use of neural, glial, mesenchymal, embryonic, and induced pluripotent stem cells. We cover current studies on stem cell replacement therapy where cells can functionally integrate into neural networks, replace damaged neurons, and strengthen impaired synaptic circuits in the brain. We address the paracrine action of stem cells acting via secreted factors to induce neuroregeneration and modify inflammatory responses. We focus on the neuroprotective functions of exosomes as well as their neurogenic and synaptogenic effects. We look into the shuttling of mitochondria through tunneling nanotubes that enables the transfer of healthy mitochondria by restoring the normal functioning of damaged cells, improving their metabolism, and reducing the level of apoptosis.
Collapse
Affiliation(s)
- Ekaterina Belousova
- Research Centre for Medical Genetics, Moscow 115522, Russia; (E.B.); (D.S.); (Y.M.); (A.S.); (D.G.)
| | - Diana Salikhova
- Research Centre for Medical Genetics, Moscow 115522, Russia; (E.B.); (D.S.); (Y.M.); (A.S.); (D.G.)
- Research Institute of Molecular and Cellular Medicine of the Medical Institute Peoples’ Friendship, University of Russia, Moscow 117198, Russia
| | - Yaroslav Maksimov
- Research Centre for Medical Genetics, Moscow 115522, Russia; (E.B.); (D.S.); (Y.M.); (A.S.); (D.G.)
- Research Institute of Molecular and Cellular Medicine of the Medical Institute Peoples’ Friendship, University of Russia, Moscow 117198, Russia
| | - Vladimir Nebogatikov
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry of the Russian Academy of Sciences, Chernogolovka 142432, Russia;
| | - Anastasiya Sudina
- Research Centre for Medical Genetics, Moscow 115522, Russia; (E.B.); (D.S.); (Y.M.); (A.S.); (D.G.)
- Research Institute of Molecular and Cellular Medicine of the Medical Institute Peoples’ Friendship, University of Russia, Moscow 117198, Russia
| | - Dmitry Goldshtein
- Research Centre for Medical Genetics, Moscow 115522, Russia; (E.B.); (D.S.); (Y.M.); (A.S.); (D.G.)
| | - Aleksey Ustyugov
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry of the Russian Academy of Sciences, Chernogolovka 142432, Russia;
| |
Collapse
|
20
|
Kukushkin NV, Carney RE, Tabassum T, Carew TJ. The massed-spaced learning effect in non-neural human cells. Nat Commun 2024; 15:9635. [PMID: 39511210 PMCID: PMC11544106 DOI: 10.1038/s41467-024-53922-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024] Open
Abstract
The massed-spaced effect is a hallmark feature of memory formation. We now demonstrate this effect in two separate non-neural, immortalized cell lines stably expressing a short-lived luciferase reporter controlled by a CREB-dependent promoter. We emulate training using repeated pulses of forskolin and/or phorbol ester, and, as a proxy for memory, measure luciferase expression at various points after training. Four spaced pulses of either agonist elicit stronger and more sustained luciferase expression than a single "massed" pulse. Spaced pulses also result in stronger and more sustained activation of molecular factors critical for memory formation, ERK and CREB, and inhibition of ERK or CREB blocks the massed-spaced effect. Our findings show that canonical features of memory do not necessarily depend on neural circuitry, but can be embedded in the dynamics of signaling cascades conserved across different cell types.
Collapse
Affiliation(s)
- N V Kukushkin
- Liberal Studies, New York University, New York, NY, 10003, USA.
- Center for Neural Science, New York University, New York, NY, 10003, USA.
| | - R E Carney
- Center for Neural Science, New York University, New York, NY, 10003, USA
| | - T Tabassum
- Center for Neural Science, New York University, New York, NY, 10003, USA
| | - T J Carew
- Center for Neural Science, New York University, New York, NY, 10003, USA.
| |
Collapse
|
21
|
Shafiek MS, Mekky RY, Nassar NN, El-Yamany MF, Rabie MA. Vortioxetine ameliorates experimental autoimmune encephalomyelitis model of multiple sclerosis in mice via activation of PI3K/Akt/CREB/BDNF cascade and modulation of serotonergic pathway signaling. Eur J Pharmacol 2024; 982:176929. [PMID: 39181226 DOI: 10.1016/j.ejphar.2024.176929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/01/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Multiple sclerosis (MS) is a chronic condition characterized by immune cell infiltration and cytokine overproduction that led to myelin sheath inflammatory assaults, thus causing axonal destruction. The former consequently provokes motor impairment and psychological disorders. Markedly, depression is one of the most prevalent lifelong comorbidities that negatively impacts the quality of life in MS patients. Vortioxetine (VTX), a multi-modal molecule prescribed to manage depression and anxiety disorder, additionally, it displays a promising neuroprotective properties against neurodegenerative diseases such as Alzheimer's and Parkinson's. To this end, the present study investigated the potential therapeutic efficacy of VTX against experimental autoimmune encephalomyelitis (EAE) model of MS in mice. Notably, treatment with VTX significantly ameliorated EAE-induced motor disability, as evident by enhanced performance in open field, rotarod and grip strength tests, alongside a reduction in immobility time during the forced swimming test, indicating a mitigation of the depressive-like behavior; outcomes that were corroborated with histological examinations and biochemical analyses. Mechanistically, VTX enhanced serotonin levels by inhibiting both serotonin transporter (SERT) and indoleamine 2,3-dioxygenase (IDO) enzyme, thereby promoting the activation of serotonin 1A (5-HT1A) receptor. The latter triggered the stimulation of phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) cascade that entailed activation/phosphorylation of cAMP response element-binding protein (CREB). This activation increased brain derived neurotrophic factor (BDNF) and myelin basic protein (MBP) contents that mitigated demyelination in the corpus callosum. Furthermore, VTX suppressed phospho serine 536 nuclear factor kappa B (pS536 NF-κB p65) activity and reduced tumor necrosis factor-alpha (TNF-α) production. The results underscore VTX's beneficial effects on disease severity in EAE model of MS in mice by amending both inflammatory and neurodegenerative components of MS progression.
Collapse
Affiliation(s)
- Marwa S Shafiek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, Egypt
| | - Radwa Y Mekky
- Department of Pharmacology and Toxicology, October University for Modern Science and Arts (MSA), Giza, 12622, Egypt
| | - Noha N Nassar
- Department of Pharmacology and Toxicology, October University for Modern Science and Arts (MSA), Giza, 12622, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| | - Mohammed F El-Yamany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| | - Mostafa A Rabie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt.
| |
Collapse
|
22
|
Zou Z, Xiao N, Chen Z, Lin X, Li Y, Li P, Cheng Q, Du B. Yeast Extract Peptides Alleviate Depression in Chronic Restraint Stress Rats by Alleviating Hippocampal Neuronal Apoptosis and Dysbiosis of the Gut Microbiota. Mol Nutr Food Res 2024; 68:e2300467. [PMID: 39432823 DOI: 10.1002/mnfr.202300467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/22/2024] [Indexed: 10/23/2024]
Abstract
SCOPE Depression as a global neurological disorder, and hippocampal neuronal apoptosis and disorders of the gut microbiota are closely related to it. This study aims to expose the ameliorative effect of enzyme peptides (AP) from brewer's yeast on depressive behavior caused by chronic restraint stress (CRS) in rats. METHODS AND RESULTS After 4 weeks of AP intervention, a significant alleviation of depressive behavior in the sucrose preference test (SPT), forced swim test (FST), and light-dark test (LDT) is observed in depressed rats. AP ameliorates neuronal damage with increased the expression of the key CREB/BDNF/TrkB/Akt signaling pathway, which increases the levels of the monoamine neurotransmitters 5-hydroxytryptamine (5-HT) and norepinephrine (NE) in the hippocampus, buffering hyperactivity of the hypothalamo-pituitary-adrenal axis (HPA), and decreasing the serum cortisol (CORT) and adrenocorticotropic hormone (ACTH) levels in rats. In addition, AP modulates the disruption of the rat gut microbiota by chronic restraint stress (CRS), and the changes in the abundance of Lactobacillus animalis and Lactobacillus johnsonii are probably the key for AP performing antidepressant benefits. A strong correlation is found between gut microbiota and biochemical markers of depression. CONCLUSION AP, as a natural and safe active substance, has a positive effect in the treatment of depression.
Collapse
Affiliation(s)
- Zebin Zou
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, 510640, China
| | - Nan Xiao
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, 510640, China
| | - Zhixian Chen
- Hubei Provincial Key Laboratory of Yeast Function, Yichang Engineering Technology Research Center of Nutrition and Health Food, Yichang, 443003, China
| | - Xucong Lin
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, 510640, China
| | - Yaqi Li
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, 510640, China
| | - Pan Li
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, 510640, China
| | - Qian Cheng
- Hubei Provincial Key Laboratory of Yeast Function, Yichang Engineering Technology Research Center of Nutrition and Health Food, Yichang, 443003, China
| | - Bing Du
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, 510640, China
| |
Collapse
|
23
|
Keady JV, Hessing MC, Songrady JC, McLaurin K, Turner JR. Sex differences in contextual fear conditioning and extinction after acute and chronic nicotine treatment. Biol Sex Differ 2024; 15:88. [PMID: 39482781 PMCID: PMC11529327 DOI: 10.1186/s13293-024-00656-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/04/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Chronic cigarette smokers report withdrawal symptomology, including affective dysfunction and cognitive deficits. While there are studies demonstrating sex specific withdrawal symptomology in nicotine-dependent individuals, literature examining the underlying biological mediators of this is scant and not in complete agreement. Therefore, in this study, we evaluated the sex specific effects of nicotine and withdrawal on contextual fear memory, a hippocampally dependent aspect of cognition that is disrupted in nicotine withdrawal. METHODS Male and female B6/129F1 mice (8-13 weeks old) were used in all experiments. For the acute nicotine experiment, mice received intraperitoneal saline or nicotine (0.5 mg/kg) prior to contextual fear conditioning and test. For the chronic nicotine experiment, mice received nicotine (18 mg/kg/day) or saline for 11 days, then underwent contextual fear conditioning and test. Following the test, mice underwent minipump removal to elicit withdrawal or sham surgery, followed by the fear extinction assay. Bulk cortical tissue was used to determine nicotinic acetylcholine receptor levels via single point [3H]Epibatidine binding assay. Gene expression levels in the dorsal and ventral hippocampus were quantified via RT-PCR. RESULTS We found that female mice had a stronger expression of contextual fear memory than their male counterparts. Further, following acute nicotine treatment, male, but not female, subjects demonstrated augmented contextual fear memory expression. In contrast, no significant effects of chronic nicotine treatment on fear conditioning were observed in either sex. When examining extinction of fear learning, we observed that female mice withdrawn from nicotine displayed impaired extinction learning, but no effect was observed in males. Nicotine withdrawal caused similar suppression of fosb, cfos, and bdnf, our proxy for neuronal activation and plasticity changes, in the dorsal and ventral hippocampus of both sexes. Additionally, we found that ventral hippocampus erbb4 expression, a gene implicated in smoking cessation outcomes, was elevated in both sexes following nicotine withdrawal. CONCLUSIONS Despite the similar impacts of nicotine withdrawal on gene expression levels, fosb, cfos, bdnf and erbb4 levels in the ventral hippocampus were predictive of delays in female extinction learning alone. This suggests sex specific dysfunction in hippocampal circuitry may contribute to female specific nicotine withdrawal induced deficits in extinction learning.
Collapse
Affiliation(s)
- Jack V Keady
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, 789 S. Limestone Street, 473 Lee T. Todd Jr. Building, Lexington, KY, 40536-0596, USA
| | - Marissa C Hessing
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, 789 S. Limestone Street, 473 Lee T. Todd Jr. Building, Lexington, KY, 40536-0596, USA
| | - Judy C Songrady
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, 789 S. Limestone Street, 473 Lee T. Todd Jr. Building, Lexington, KY, 40536-0596, USA
| | - Kristen McLaurin
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, 789 S. Limestone Street, 473 Lee T. Todd Jr. Building, Lexington, KY, 40536-0596, USA
| | - Jill R Turner
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, 789 S. Limestone Street, 473 Lee T. Todd Jr. Building, Lexington, KY, 40536-0596, USA.
| |
Collapse
|
24
|
Fukuchi M, Shibasaki Y, Akazawa Y, Suzuki-Masuyama H, Takeuchi KI, Iwazaki Y, Tabuchi A, Tsuda M. Neuron-selective and activity-dependent splicing of BDNF exon I-IX pre-mRNA. Neurochem Int 2024; 181:105889. [PMID: 39455010 DOI: 10.1016/j.neuint.2024.105889] [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: 06/27/2024] [Revised: 10/04/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
Brain-derived neurotrophic factor (BDNF) is essential for numerous neuronal functions, including learning and memory. The expression of BDNF is regulated by distinctive transcriptional and post-transcriptional mechanisms. The Bdnf gene in mice and rats comprises eight untranslated exons (exons I-VIII) and one exon (exon IX) that contains the pre-proBDNF coding sequence. Multiple splice donor sites on the untranslated exons and a single acceptor site upstream of the coding sequence result in the characteristic exon skipping patterns that generate multiple Bdnf mRNA variants, which are essential for the spatiotemporal regulation of BDNF expression, mRNA localization, mRNA stability, and translational control. However, the regulation of Bdnf pre-mRNA splicing remains unclear. Here, we focused on the splicing of Bdnf exon I-IX pre-mRNA. We first constructed a minigene to evaluate Bdnf exon I-IX pre-mRNA splicing. Compared with Bdnf exon I-IX pre-mRNA splicing in non-neuronal NIH3T3 cells, splicing was preferentially observed in primary cultures of cortical neurons. Additionally, a series of overexpression and knockdown experiments suggested that neuro-oncological ventral antigen (NOVA) 2 is involved in the neuron-selective splicing of Bdnf exon I-IX pre-mRNA. Supporting this finding, endogenous Nova2 mRNA expression was markedly higher in neurons, and a strong correlation between endogenous Bdnf exon I-IX and Nova2 mRNA was observed across several brain regions. Furthermore, Bdnf exon I-IX pre-mRNA splicing was facilitated by Ca2+ signals evoked via L-type voltage-dependent Ca2+ channels. Notably, among the Bdnf pre-mRNA splicing investigated in the current study, neuron-selective and activity-dependent splicing was observed in Bdnf exon I-IX pre-mRNA. In conclusion, Bdnf exon I-IX pre-mRNA splicing is preferentially observed in neurons and is facilitated in an activity-dependent manner. The neuron-selective and activity-dependent splicing of Bdnf exon I-IX pre-mRNA may contribute to the efficient induction of Bdnf exon I-IX expression in neurons.
Collapse
Affiliation(s)
- Mamoru Fukuchi
- Laboratory of Molecular Neuroscience, Faculty of Pharmacy, Takasaki University of Health and Welfare, 60 Nakaorui-machi, Takasaki, Gunma, 370-0033, Japan; Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.
| | - Yumi Shibasaki
- Laboratory of Molecular Neuroscience, Faculty of Pharmacy, Takasaki University of Health and Welfare, 60 Nakaorui-machi, Takasaki, Gunma, 370-0033, Japan
| | - Yuto Akazawa
- Laboratory of Molecular Neuroscience, Faculty of Pharmacy, Takasaki University of Health and Welfare, 60 Nakaorui-machi, Takasaki, Gunma, 370-0033, Japan
| | - Hitoshi Suzuki-Masuyama
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Ken-Ichi Takeuchi
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Yumika Iwazaki
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Akiko Tabuchi
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Masaaki Tsuda
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| |
Collapse
|
25
|
Katimbwa DA, Kim Y, Kim MJ, Jeong M, Lim J. Solubilized β-Glucan Supplementation in C57BL/6J Mice Dams Augments Neurodevelopment and Cognition in the Offspring Driven by Gut Microbiome Remodeling. Foods 2024; 13:3102. [PMID: 39410136 PMCID: PMC11476385 DOI: 10.3390/foods13193102] [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: 09/09/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
A maternal diet rich in dietary fiber, such as β-glucan, plays a crucial role in the offspring's acquisition of gut microbiota and the subsequent shaping of its microbiome profile and metabolome. This in turn has been shown to aid in neurodevelopmental processes, including early microglial maturation and immunomodulation via metabolites like short chain fatty acids (SCFAs). This study aimed to investigate the effects of oat β-glucan supplementation, solubilized by citric acid hydrolysis, from gestation to adulthood. Female C57BL/6J mice were orally supplemented with soluble oat β-glucan (ObG) or carboxymethyl cellulose (CMC) via drinking water at 200 mg/kg body weight during breeding while the control group received 50 mg/kg body weight of carboxymethyl cellulose. ObG supplementation increased butyrate production in the guts of both dams and 4-week-old pups, attributing to alterations in the gut microbiota profile. One-week-old pups from the ObG group showed increased neurodevelopmental markers similar to four-week-old pups that also exhibited alterations in serum markers of metabolism and anti-inflammatory cytokines. Notably, at 8 weeks, ObG-supplemented pups exhibited the highest levels of spatial memory and cognition compared to the control and CMC groups. These findings suggest a potential enhancement of neonatal neurodevelopment via shaping of early-life gut microbiome profile, and the subsequent increased later-life cognitive function.
Collapse
Affiliation(s)
- Dorsilla A. Katimbwa
- Department of Food Biomaterials, Kyungpook National University, Daegu 41566, Republic of Korea;
| | - Yoonsu Kim
- Department of Integrative Biology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Min Jeong Kim
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Minsoo Jeong
- Department of Applied Biosciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jinkyu Lim
- Department of Food Biomaterials, Kyungpook National University, Daegu 41566, Republic of Korea;
| |
Collapse
|
26
|
Chowdhury MAR, Haq MM, Lee JH, Jeong S. Multi-faceted regulation of CREB family transcription factors. Front Mol Neurosci 2024; 17:1408949. [PMID: 39165717 PMCID: PMC11333461 DOI: 10.3389/fnmol.2024.1408949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 07/12/2024] [Indexed: 08/22/2024] Open
Abstract
cAMP response element-binding protein (CREB) is a ubiquitously expressed nuclear transcription factor, which can be constitutively activated regardless of external stimuli or be inducibly activated by external factors such as stressors, hormones, neurotransmitters, and growth factors. However, CREB controls diverse biological processes including cell growth, differentiation, proliferation, survival, apoptosis in a cell-type-specific manner. The diverse functions of CREB appear to be due to CREB-mediated differential gene expression that depends on cAMP response elements and multi-faceted regulation of CREB activity. Indeed, the transcriptional activity of CREB is controlled at several levels including alternative splicing, post-translational modification, dimerization, specific transcriptional co-activators, non-coding small RNAs, and epigenetic regulation. In this review, we present versatile regulatory modes of CREB family transcription factors and discuss their functional consequences.
Collapse
Affiliation(s)
- Md Arifur Rahman Chowdhury
- Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju, Republic of Korea
- Department of Molecular Biology, and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Republic of Korea
| | - Md Mazedul Haq
- Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju, Republic of Korea
- Department of Molecular Biology, and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Republic of Korea
| | - Jeong Hwan Lee
- Division of Life Sciences, Jeonbuk National University, Jeonju, Republic of Korea
| | - Sangyun Jeong
- Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju, Republic of Korea
- Department of Molecular Biology, and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Republic of Korea
| |
Collapse
|
27
|
Wolf D, Ayon-Olivas M, Sendtner M. BDNF-Regulated Modulation of Striatal Circuits and Implications for Parkinson's Disease and Dystonia. Biomedicines 2024; 12:1761. [PMID: 39200225 PMCID: PMC11351984 DOI: 10.3390/biomedicines12081761] [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: 07/02/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 09/02/2024] Open
Abstract
Neurotrophins, particularly brain-derived neurotrophic factor (BDNF), act as key regulators of neuronal development, survival, and plasticity. BDNF is necessary for neuronal and functional maintenance in the striatum and the substantia nigra, both structures involved in the pathogenesis of Parkinson's Disease (PD). Depletion of BDNF leads to striatal degeneration and defects in the dendritic arborization of striatal neurons. Activation of tropomyosin receptor kinase B (TrkB) by BDNF is necessary for the induction of long-term potentiation (LTP), a form of synaptic plasticity, in the hippocampus and striatum. PD is characterized by the degeneration of nigrostriatal neurons and altered striatal plasticity has been implicated in the pathophysiology of PD motor symptoms, leading to imbalances in the basal ganglia motor pathways. Given its essential role in promoting neuronal survival and meditating synaptic plasticity in the motor system, BDNF might have an important impact on the pathophysiology of neurodegenerative diseases, such as PD. In this review, we focus on the role of BDNF in corticostriatal plasticity in movement disorders, including PD and dystonia. We discuss the mechanisms of how dopaminergic input modulates BDNF/TrkB signaling at corticostriatal synapses and the involvement of these mechanisms in neuronal function and synaptic plasticity. Evidence for alterations of BDNF and TrkB in PD patients and animal models are reviewed, and the potential of BDNF to act as a therapeutic agent is highlighted. Advancing our understanding of these mechanisms could pave the way toward innovative therapeutic strategies aiming at restoring neuroplasticity and enhancing motor function in these diseases.
Collapse
Affiliation(s)
| | | | - Michael Sendtner
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, 97078 Wuerzburg, Germany (M.A.-O.)
| |
Collapse
|
28
|
Inagaki R, Yamakuni T, Saito T, Saido TC, Moriguchi S. Preventive effect of propolis on cognitive decline in Alzheimer's disease model mice. Neurobiol Aging 2024; 139:20-29. [PMID: 38583392 DOI: 10.1016/j.neurobiolaging.2024.03.002] [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: 12/05/2023] [Revised: 02/20/2024] [Accepted: 03/11/2024] [Indexed: 04/09/2024]
Abstract
Brazilian green propolis (propolis) is a chemically complex resinous substance that is a potentially viable therapeutic agent for Alzheimer's disease. Herein, propolis induced a transient increase in intracellular Ca2+ concentration ([Ca2+]i) in Neuro-2A cells; moreover, propolis-induced [Ca2+]i elevations were suppressed prior to 24-h pretreatment with amyloid-β. To reveal the effect of [Ca2+]i elevation on impaired cognition, we performed memory-related behavioral tasks in APP-KI mice relative to WT mice at 4 and 12 months of age. Propolis, at 300-1000 mg/kg/d for 8 wk, significantly ameliorated cognitive deficits in APP-KI mice at 4 months, but not at 12 months of age. Consistent with behavioral observations, injured hippocampal long-term potentiation was markedly ameliorated in APP-KI mice at 4 months of age following repeated propolis administration. In addition, repeated administration of propolis significantly activated intracellular calcium signaling pathway in the CA1 region of APP-KI mice. These results suggest a preventive effect of propolis on cognitive decline through the activation of intracellular calcium signaling pathways in CA1 region of AD mice model.
Collapse
Affiliation(s)
- Ryo Inagaki
- Research Center for Pharmaceutical Development, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Tohru Yamakuni
- Research Center of Supercritical Fluid Technology, Graduate School of Engineering, Tohoku University, Sendai, Japan; New Industry Creation Hatchery Center, Tohoku University, Sendai, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Saitama, Japan
| | - Shigeki Moriguchi
- Research Center for Pharmaceutical Development, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
| |
Collapse
|
29
|
Bastos CR, Bevilacqua LM, Mendes LFB, Xavier J, Gruhn K, Kaster MP, Ghisleni G. Amygdala-specific changes in Cacna1c, Nfat5, and Bdnf expression are associated with stress responsivity in mice: A possible mechanism for psychiatric disorders. J Psychiatr Res 2024; 175:259-270. [PMID: 38754148 DOI: 10.1016/j.jpsychires.2024.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 03/11/2024] [Accepted: 05/07/2024] [Indexed: 05/18/2024]
Abstract
The CACNA1C gene encodes the alpha-1c subunit of the Cav1.2 calcium channel, a regulator of neuronal calcium influx involved in neurotransmitter release and synaptic plasticity. Genetic data show a role for CACNA1C in depressive symptoms underlying different psychiatric diagnoses. However, the mechanisms involved still require further exploration. This study aimed to investigate sex and region-specific changes in the Cacna1c gene and behavioral outcomes in mice exposed to chronic stress. Moreover, we evaluated the Nuclear factor of activated T-cells 5 (Nfat5) and the Brain-derived neurotrophic factor (Bdnf) as potential upstream and downstream Cacna1c targets and their correlation in stressed mice and humans with depression. Male and female Swiss mice were exposed to chronic unpredictable stress (CUS) for 21 days. Animal-integrated emotionality was assessed using the sucrose splash test, the tail suspension, the open-field test, and the elevated-plus-maze. Gene expression analysis was performed in the amygdala, prefrontal cortex, and hippocampus. Human data for in silico analysis was obtained from the Gene Expression Omnibus. CUS-induced impairment in integrated emotional regulation was observed in males. Gene expression analysis showed decreased levels of Cacna1c and Nfat5 and increased levels of Bdnf transcripts in the amygdala of stressed male mice. In contrast, there were no major changes in behavioral responses or gene expression in female mice after stress. The expression of the three genes was significantly correlated in the amygdala of mice and humans. The strong and positive correlation between Canac1c and Nfat5 suggests a potential role for this transcription factor in Canac1c expression. These changes could impact amygdala reactivity and emotional responses, making them a potential target for psychiatric intervention.
Collapse
Affiliation(s)
- Clarissa Ribeiro Bastos
- Laboratory of Translational Neuroscience, Department of Biochemistry, Federal University of Santa Catarina (UFSC), Florianopolis, Santa Catarina, Brazil; Department of Life and Health Sciences, Catholic University of Pelotas (UCPel), Pelotas, Rio Grande do Sul, Brazil
| | - Laura Menegatti Bevilacqua
- Laboratory of Translational Neuroscience, Department of Biochemistry, Federal University of Santa Catarina (UFSC), Florianopolis, Santa Catarina, Brazil
| | - Luiz Filipe Bastos Mendes
- Center of Oxidative Stress Research, Department of Biochemistry, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Janaina Xavier
- Department of Life and Health Sciences, Catholic University of Pelotas (UCPel), Pelotas, Rio Grande do Sul, Brazil
| | - Karen Gruhn
- Department of Life and Health Sciences, Catholic University of Pelotas (UCPel), Pelotas, Rio Grande do Sul, Brazil
| | - Manuella Pinto Kaster
- Laboratory of Translational Neuroscience, Department of Biochemistry, Federal University of Santa Catarina (UFSC), Florianopolis, Santa Catarina, Brazil.
| | - Gabriele Ghisleni
- Department of Life and Health Sciences, Catholic University of Pelotas (UCPel), Pelotas, Rio Grande do Sul, Brazil.
| |
Collapse
|
30
|
Liu MN, Lan Q, Wu H, Qiu CW. Rejuvenation of young blood on aging organs: Effects, circulating factors, and mechanisms. Heliyon 2024; 10:e32652. [PMID: 38994040 PMCID: PMC11237939 DOI: 10.1016/j.heliyon.2024.e32652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 06/06/2024] [Indexed: 07/13/2024] Open
Abstract
Aging causes degenerative changes in organs, leading to a decline in physical function. Over the past two decades, researchers have made significant progress in understanding the rejuvenating effects of young blood on aging organs, benefiting from heterochronic parabiosis models that connect the blood circulation of aged and young rodents. It has been discovered that young blood can partially rejuvenate organs in old animals by regulating important aging-related signaling pathways. Clinical trials have also shown the effectiveness of young blood in treating aging-related diseases. However, the limited availability of young blood poses a challenge to implementing anti-aging therapies on a large scale for older individuals. As a promising alternative, scientists have identified some specific anti-aging circulating factors in young blood that have been shown to promote organ regeneration, reduce inflammation, and alleviate fibrosis associated with aging in animal experiments. While previous reviews have focused primarily on the effects and mechanisms of circulating factors on aging, it is important to acknowledge that studying the rejuvenating effects and mechanisms of young blood has been a significant source of inspiration in this field, and it will continue to be in the future. In recent years, new findings have emerged, further expanding our knowledge in this area. This review aims to summarize the rejuvenating effects and mechanisms of young blood and circulating factors, discussing their similarities and connections, addressing discrepancies in previous studies, outlining future research directions, and highlighting the potential for clinical translation in anti-aging interventions.
Collapse
Affiliation(s)
- Meng-Nan Liu
- National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, PR China
| | - Qi Lan
- National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, PR China
| | - Hao Wu
- National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, PR China
| | - Cai-Wei Qiu
- Research Center of Combine Traditional Chinese and Western Medicine, Prophylaxis and Treatment of Organ Fibrosis by Integrated Medicine of Luzhou Key Laboratory, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, PR China
| |
Collapse
|
31
|
Drexler R, Khatri R, Sauvigny T, Mohme M, Maire CL, Ryba A, Zghaibeh Y, Dührsen L, Salviano-Silva A, Lamszus K, Westphal M, Gempt J, Wefers AK, Neumann JE, Bode H, Hausmann F, Huber TB, Bonn S, Jütten K, Delev D, Weber KJ, Harter PN, Onken J, Vajkoczy P, Capper D, Wiestler B, Weller M, Snijder B, Buck A, Weiss T, Göller PC, Sahm F, Menstel JA, Zimmer DN, Keough MB, Ni L, Monje M, Silverbush D, Hovestadt V, Suvà ML, Krishna S, Hervey-Jumper SL, Schüller U, Heiland DH, Hänzelmann S, Ricklefs FL. A prognostic neural epigenetic signature in high-grade glioma. Nat Med 2024; 30:1622-1635. [PMID: 38760585 PMCID: PMC11186787 DOI: 10.1038/s41591-024-02969-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 04/03/2024] [Indexed: 05/19/2024]
Abstract
Neural-tumor interactions drive glioma growth as evidenced in preclinical models, but clinical validation is limited. We present an epigenetically defined neural signature of glioblastoma that independently predicts patients' survival. We use reference signatures of neural cells to deconvolve tumor DNA and classify samples into low- or high-neural tumors. High-neural glioblastomas exhibit hypomethylated CpG sites and upregulation of genes associated with synaptic integration. Single-cell transcriptomic analysis reveals a high abundance of malignant stemcell-like cells in high-neural glioblastoma, primarily of the neural lineage. These cells are further classified as neural-progenitor-cell-like, astrocyte-like and oligodendrocyte-progenitor-like, alongside oligodendrocytes and excitatory neurons. In line with these findings, high-neural glioblastoma cells engender neuron-to-glioma synapse formation in vitro and in vivo and show an unfavorable survival after xenografting. In patients, a high-neural signature is associated with decreased overall and progression-free survival. High-neural tumors also exhibit increased functional connectivity in magnetencephalography and resting-state magnet resonance imaging and can be detected via DNA analytes and brain-derived neurotrophic factor in patients' plasma. The prognostic importance of the neural signature was further validated in patients diagnosed with diffuse midline glioma. Our study presents an epigenetically defined malignant neural signature in high-grade gliomas that is prognostically relevant. High-neural gliomas likely require a maximized surgical resection approach for improved outcomes.
Collapse
Affiliation(s)
- Richard Drexler
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Neurology, Stanford University, Stanford, CA, USA
| | - Robin Khatri
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Sauvigny
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malte Mohme
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cecile L Maire
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alice Ryba
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yahya Zghaibeh
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lasse Dührsen
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Amanda Salviano-Silva
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katrin Lamszus
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manfred Westphal
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jens Gempt
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Annika K Wefers
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julia E Neumann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Center for Molecular Neurobiology Hamburg (ZMNH), University Hospital Hamburg Eppendorf, Hamburg, Germany
| | - Helena Bode
- Research Institute Children's Cancer Center Hamburg, Hamburg, Germany
| | - Fabian Hausmann
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Bonn
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kerstin Jütten
- Department of Neurosurgery, University Hospital Aachen, Aachen, Germany
| | - Daniel Delev
- Department of Neurosurgery, University Hospital Aachen, Aachen, Germany
- Department of Neurosurgery, University Clinic Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Katharina J Weber
- Neurological Institute (Edinger Institute), University Hospital Frankfurt, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
- University Cancer Center (UCT) Frankfurt, Frankfurt am Main, Germany
| | - Patrick N Harter
- Neurological Institute (Edinger Institute), University Hospital Frankfurt, Frankfurt am Main, Germany
- Institute of Neuropathology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Julia Onken
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - David Capper
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Benedikt Wiestler
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University Munich, Munich, Germany
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich, Zurich, Switzerland
- Department of Neurology, University of Zürich, Zurich, Switzerland
| | - Berend Snijder
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Alicia Buck
- Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich, Zurich, Switzerland
- Department of Neurology, University of Zürich, Zurich, Switzerland
| | - Tobias Weiss
- Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich, Zurich, Switzerland
- Department of Neurology, University of Zürich, Zurich, Switzerland
| | - Pauline C Göller
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Felix Sahm
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Joelle Aline Menstel
- Department of Neurosurgery, Medical Center University of Freiburg, Freiburg, Germany
| | - David Niklas Zimmer
- Department of Neurosurgery, Medical Center University of Freiburg, Freiburg, Germany
| | | | - Lijun Ni
- Department of Neurology, Stanford University, Stanford, CA, USA
| | - Michelle Monje
- Department of Neurology, Stanford University, Stanford, CA, USA
| | - Dana Silverbush
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Volker Hovestadt
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Mario L Suvà
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Saritha Krishna
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Shawn L Hervey-Jumper
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Ulrich Schüller
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children's Cancer Center Hamburg, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, Research Institute Children's Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dieter H Heiland
- Department of Neurosurgery, University Clinic Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Neurosurgery, Medical Center University of Freiburg, Freiburg, Germany
- Translational Neurosurgery, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Sonja Hänzelmann
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Franz L Ricklefs
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
32
|
Lee HW, Chen SJ, Tsai KJ, Hsu KS, Chen YF, Chang CH, Lin HH, Hsueh WY, Hsieh HP, Lee YF, Chiang HC, Chang JY. Targeting cathepsin S promotes activation of OLF1-BDNF/TrkB axis to enhance cognitive function. J Biomed Sci 2024; 31:46. [PMID: 38725007 PMCID: PMC11084077 DOI: 10.1186/s12929-024-01037-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 04/27/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Cathepsin S (CTSS) is a cysteine protease that played diverse roles in immunity, tumor metastasis, aging and other pathological alterations. At the cellular level, increased CTSS levels have been associated with the secretion of pro-inflammatory cytokines and disrupted the homeostasis of Ca2+ flux. Once CTSS was suppressed, elevated levels of anti-inflammatory cytokines and changes of Ca2+ influx were observed. These findings have inspired us to explore the potential role of CTSS on cognitive functions. METHODS We conducted classic Y-maze and Barnes Maze tests to assess the spatial and working memory of Ctss-/- mice, Ctss+/+ mice and Ctss+/+ mice injected with the CTSS inhibitor (RJW-58). Ex vivo analyses including long-term potentiation (LTP), Golgi staining, immunofluorescence staining of sectioned whole brain tissues obtained from experimental animals were conducted. Furthermore, molecular studies were carried out using cultured HT-22 cell line and primary cortical neurons that treated with RJW-58 to comprehensively assess the gene and protein expressions. RESULTS Our findings reported that targeting cathepsin S (CTSS) yields improvements in cognitive function, enhancing both working and spatial memory in behavior models. Ex vivo studies showed elevated levels of long-term potentiation levels and increased synaptic complexity. Microarray analysis demonstrated that brain-derived neurotrophic factor (BDNF) was upregulated when CTSS was knocked down by using siRNA. Moreover, the pharmacological blockade of the CTSS enzymatic activity promoted BDNF expression in a dose- and time-dependent manner. Notably, the inhibition of CTSS was associated with increased neurogenesis in the murine dentate gyrus. These results suggested a promising role of CTSS modulation in cognitive enhancement and neurogenesis. CONCLUSION Our findings suggest a critical role of CTSS in the regulation of cognitive function by modulating the Ca2+ influx, leading to enhanced activation of the BDNF/TrkB axis. Our study may provide a novel strategy for improving cognitive function by targeting CTSS.
Collapse
Affiliation(s)
- Hao-Wei Lee
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
- Taipei Cancer Center, TMU Research Center of Cancer Translational Medicine, Taipei Medical University Hospital, College of Medicine, Taipei Medical University, No. 252, Wuxing St., Xinyi Dist., Taipei, 110301, Taiwan (R.O.C.)
| | - Szu-Jung Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Kuen-Jer Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Research Center of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kuei-Sen Hsu
- Institute of Basic Medical Science, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Fan Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Chih-Hua Chang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Hsiao-Han Lin
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Wen-Yun Hsueh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Hsing-Pang Hsieh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Yueh-Feng Lee
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Huai-Chueh Chiang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Jang-Yang Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan.
- Taipei Cancer Center, TMU Research Center of Cancer Translational Medicine, Taipei Medical University Hospital, College of Medicine, Taipei Medical University, No. 252, Wuxing St., Xinyi Dist., Taipei, 110301, Taiwan (R.O.C.).
| |
Collapse
|
33
|
Meuten TK, Dean GA, Thamm DH. Review: The PI3K-AKT-mTOR signal transduction pathway in canine cancer. Vet Pathol 2024; 61:339-356. [PMID: 37905509 DOI: 10.1177/03009858231207021] [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: 11/02/2023]
Abstract
Tumors in dogs and humans share many similar molecular and genetic features, incentivizing a better understanding of canine neoplasms not only for the purpose of treating companion animals, but also to facilitate research of spontaneously developing tumors with similar biologic behavior and treatment approaches in an immunologically competent animal model. Multiple tumor types of both species have similar dysregulation of signal transduction through phosphatidylinositol 3-kinase (PI3K), protein kinase B (PKB; AKT), and mechanistic target of rapamycin (mTOR), collectively known as the PI3K-AKT-mTOR pathway. This review aims to delineate the pertinent aspects of the PI3K-AKT-mTOR signaling pathway in health and in tumor development. It will then present a synopsis of current understanding of PI3K-AKT-mTOR signaling in important canine cancers and advancements in targeted inhibitors of this pathway.
Collapse
|
34
|
Lv D, Xiao B, Liu H, Wang L, Li Y, Zhang YH, Jin Q. Enhanced NMDA receptor pathway and glutamate transmission in the hippocampal dentate gyrus mediate the spatial learning and memory impairment of obese rats. Pflugers Arch 2024; 476:821-831. [PMID: 38416255 PMCID: PMC11033237 DOI: 10.1007/s00424-024-02924-1] [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: 10/04/2023] [Revised: 01/30/2024] [Accepted: 02/07/2024] [Indexed: 02/29/2024]
Abstract
Obesity has been linked with the impairment of spatial memory and synaptic plasticity but the molecular mechanisms remained unidentified. Since glutamatergic transmission and NMDA receptor neural pathways in hippocampal dentate gyrus (DG) are essential in the learning and memory, we aimed to investigate glutamate (Glu) and NMDA receptor signaling of DG in spatial learning and memory in diet-induced obesity (DIO) rats. Spatial learning and memory were assessed via Morris water maze (MWM) test on control (Ctr) and DIO rats. Extracellular concentration of Glu in the DG was determined using in vivo microdialysis and HPLC. The protein expressions of NMDA receptor subunit 2B (NR2B), brain-derived neurotrophic factor (BDNF), the activation of calcium/calmodulin-dependent kinase II (CaMKII) and cAMP-response-element-binding protein (CREB) in the DG were observed by western blot. Spatial learning and memory were impaired in DIO rats compared to those of Ctr. NR2B expression was increased, while BDNF expression and CaMKII and CREB activation were decreased in DG of DIO rats. Extracellular concentration of Glu was increased in Ctr on the 3rd and 4th days of the MWM test, but significant further increment was observed in DIO rats. Microinjection of an NMDA antagonist (MK-801) into the DG reversed spatial learning and memory impairment. Such effects were accompanied by greater BDNF expression and CaMKII/CREB activation in the DG of DIO rats. In conclusion, the enhancement of Glu-NMDA receptor transmission in the hippocampal DG contributes to the impairment of spatial learning and memory in DIO rats, maybe via the modulation of CaMKII-CREB-BDNF signaling pathway.
Collapse
Affiliation(s)
- Dingding Lv
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, 133002, China
| | - Bin Xiao
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, 133002, China
| | - Huaying Liu
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, 133002, China
| | - Linping Wang
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, 133002, China
| | - Yingshun Li
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, 133002, China
| | - Yin Hua Zhang
- Department of Physiology and Biomedical Sciences, Ischemia/Hypoxic Disease Institute, Seoul National University, College of Medicine, Seoul, Korea.
| | - Qinghua Jin
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, 133002, China.
| |
Collapse
|
35
|
Liu S, Lu Y, Tian D, Zhang T, Zhang C, Hu CY, Chen P, Meng Y. Hydroxytyrosol Alleviates Obesity-Induced Cognitive Decline by Modulating the Expression Levels of Brain-Derived Neurotrophic Factors and Inflammatory Factors in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:6250-6264. [PMID: 38491001 DOI: 10.1021/acs.jafc.3c08319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2024]
Abstract
Hydroxytyrosol (HT; 3,4-dihydroxyphenyl ethanol) is an important functional polyphenol in olive oil. Our study sought to evaluate the protective effects and underlying mechanisms of HT on obesity-induced cognitive impairment. A high-fat and high-fructose-diet-induced obese mice model was treated with HT for 14 weeks. The results show that HT improved the learning and memory abilities and enhanced the expressions of brain-derived neurotrophic factors (BDNFs) and postsynaptic density proteins, protecting neuronal and synaptic functions in obese mice. Transcriptomic results further confirmed that HT improved cognitive impairment by regulating gene expression in neural system development and synaptic function-related pathways. Moreover, HT treatment alleviated neuroinflammation in the brain of obese mice. To sum up, our results indicated that HT can alleviate obesity-induced cognitive dysfunction by enhancing BDNF expression and alleviating neuroinflammation in the brain, which also means that HT may become a potentially useful nutritional supplement to alleviate obesity-induced cognitive decline.
Collapse
Affiliation(s)
- Shenlin Liu
- The Engineering Research Center of High-Valued Utilization of Fruit Resources in Western China, Ministry of Education; National Research & Development Center of Apple Processing Technology; College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, P. R. China
| | - Yalong Lu
- The Engineering Research Center of High-Valued Utilization of Fruit Resources in Western China, Ministry of Education; National Research & Development Center of Apple Processing Technology; College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, P. R. China
| | - Dan Tian
- The Engineering Research Center of High-Valued Utilization of Fruit Resources in Western China, Ministry of Education; National Research & Development Center of Apple Processing Technology; College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, P. R. China
| | - Tingting Zhang
- The Engineering Research Center of High-Valued Utilization of Fruit Resources in Western China, Ministry of Education; National Research & Development Center of Apple Processing Technology; College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, P. R. China
| | - Chaoqun Zhang
- The Engineering Research Center of High-Valued Utilization of Fruit Resources in Western China, Ministry of Education; National Research & Development Center of Apple Processing Technology; College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, P. R. China
| | - Ching Yuan Hu
- The Engineering Research Center of High-Valued Utilization of Fruit Resources in Western China, Ministry of Education; National Research & Development Center of Apple Processing Technology; College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, P. R. China
- Department of Human Nutrition, Food and Animal Sciences, College of Tropical Agriculture and Human Resources, University of Hawaii at Manoa, 1955 East-West Road, AgSci. 415J, Honolulu, Hawaii 96822, United States
| | - Ping Chen
- Shaanxi Provincial Center for Disease Control and Prevention, Xian, Shaanxi 710054, P. R. China
| | - Yonghong Meng
- The Engineering Research Center of High-Valued Utilization of Fruit Resources in Western China, Ministry of Education; National Research & Development Center of Apple Processing Technology; College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, P. R. China
| |
Collapse
|
36
|
Albin B, Adhikari P, Tiwari AP, Qubbaj K, Yang IH. Electrical stimulation enhances mitochondrial trafficking as a neuroprotective mechanism against chemotherapy-induced peripheral neuropathy. iScience 2024; 27:109052. [PMID: 38375222 PMCID: PMC10875116 DOI: 10.1016/j.isci.2024.109052] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/20/2023] [Accepted: 01/23/2024] [Indexed: 02/21/2024] Open
Abstract
Electrical stimulation (ESTIM) has shown to be an effective symptomatic treatment to treat pain associated with peripheral nerve damage. However, the neuroprotective mechanism of ESTIM on peripheral neuropathies is still unknown. In this study, we identified that ESTIM has the ability to enhance mitochondrial trafficking as a neuroprotective mechanism against chemotherapy-induced peripheral neuropathies (CIPNs). CIPN is a debilitating and painful sequalae of anti-cancer chemotherapy treatment which results in degeneration of peripheral nerves. Mitochondrial dynamics were analyzed within axons in response to two different antineoplastic mechanisms by chemotherapy drug treatments paclitaxel and oxaliplatin in vitro. Mitochondrial trafficking response to chemotherapy drug treatment was observed to decrease in conjunction with degeneration of distal axons. Using low-frequency ESTIM, we observed enhanced mitochondrial trafficking to be a neuroprotective mechanism against CIPN. This study confirms ESTIM enhances regeneration of peripheral nerves by increased mitochondrial trafficking.
Collapse
Affiliation(s)
- Bayne Albin
- Center for Biomedical Engineering and Science, Department of Mechanical Engineering and Engineering Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Prashant Adhikari
- Center for Biomedical Engineering and Science, Department of Mechanical Engineering and Engineering Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Arjun Prasad Tiwari
- Center for Biomedical Engineering and Science, Department of Mechanical Engineering and Engineering Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Khayzaran Qubbaj
- Center for Biomedical Engineering and Science, Department of Mechanical Engineering and Engineering Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - In Hong Yang
- Center for Biomedical Engineering and Science, Department of Mechanical Engineering and Engineering Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| |
Collapse
|
37
|
Sims SK, Saddow M, McGonegal L, Sims-Robinson C. Intranasal Administration of BDNF Improves Recovery and Promotes Neural Plasticity in a Neonatal Mouse Model of Hypoxic Ischemia. Exp Neurobiol 2024; 33:25-35. [PMID: 38471802 PMCID: PMC10938072 DOI: 10.5607/en23030] [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: 09/02/2023] [Revised: 01/18/2024] [Accepted: 02/09/2024] [Indexed: 03/14/2024] Open
Abstract
The benefit of intranasal brain derived neurotrophic factor (BDNF) treatment on cognitive function in a neonatal postnatal day 7 (P7) mouse model of hypoxic ischemia (HI) was explored. Intranasal delivery is attractive in that it can promote widespread distribution of BDNF within both the brain and spinal cord. In this study we evaluated the effectiveness of intranasal BDNF to improve cognitive recovery following HI. HI is induced via ligation of the right carotid artery followed by a 45-minute exposure to an 8% oxygen/ 92% nitrogen mixture in an enclosed chamber. Male and female pups were subjected to a 2-hour hypothermia in a temperature-controlled chamber as a standard of care. A solution of saline (control) or recombinant human BDNF (Harlan Laboratories) was administered with a Gilson pipette at the same time each day for 7 days into each nasal cavity in awake mice beginning 24 hours after HI. We evaluated cognitive recovery using the novel object recognition (NOR) and western analysis to analyze neuro-markers and brain health such as synaptophysin and microtubule associated protein -2 (MAP2). The objective of this study was to evaluate the role and therapeutic potential of BDNF in neonatal HI recovery. Our results indicate that intranasal BDNF delivered within 24 hours after HI improved object discrimination at both 28 and 42 days after HI. Our results also demonstrate increased synaptophysin and MAP2 at day 42 in HI animals that received intranasal BDNF treatment compared to HI animals that were administered saline.
Collapse
Affiliation(s)
- Serena-Kaye Sims
- Department of Neurology, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Biology, College of Charleston, Charleston, SC 29424, USA
| | - Madelynne Saddow
- Department of Neurology, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Biology, College of Charleston, Charleston, SC 29424, USA
| | - Lilly McGonegal
- Department of Neurology, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Biology, College of Charleston, Charleston, SC 29424, USA
| | - Catrina Sims-Robinson
- Department of Neurology, Medical University of South Carolina, Charleston, SC 29425, USA
- Ralph H Johnson Veterans Affairs Medical Center, Charleston, SC 29401, USA
| |
Collapse
|
38
|
Zhang Y, Wang D, Liu J, Sun J, Liu X, Fan B, Lu C, Wang F. Investigating the Antidepressant Mechanisms of Polygonum sibiricum Polysaccharides via Microglial Polarization. Nutrients 2024; 16:438. [PMID: 38337722 PMCID: PMC10856971 DOI: 10.3390/nu16030438] [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: 12/20/2023] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Polygonum sibiricum, with its medicinal and edibility dual properties, has been widely recognized and utilized throughout Chinese history. As a kind of its effective component, Polygonum sibiricum polysaccharides (PSP) have been reported to be a promising novel antidepressant agent. Meanwhile, the precise mechanisms underlying its action remain elusive. The polarization state transition of microglia is intricately linked to neuroinflammation, indicating its crucial involvement in the pathophysiology of depression. Researchers are vigorously pursuing the exploration of this potential treatment strategy, aiming to comprehend its underlying mechanisms. Hence, the current study was designed to investigate the antidepressant mechanisms of PSP via Microglial M1/M2 Polarization, based on the lipopolysaccharide (LPS)-induced BV2 cell activation model. The results indicate that PSP significantly inhibited NO and LDH release and reduced ROS levels in LPS-induced BV2 cells. PSP could significantly reduce the protein expression level of Iba-1, decreased the mRNA levels of TNF-α, IL-1β, and IL-6, and increased the mRNA level of IL-10. PSP also significantly reduced the protein expression level of CD16/32 and increased that of CD206, reduced the mRNA level and fluorescence intensity of iNOS, and increased those of Arg-1. However, PSP pretreatment reversed the alterations of the BDNF/TrkB/CREB and Notch/Hes1 pathways in LPS-induced BV2 cells. These results suggested that PSP exerted the anti-inflammatory effects by inhibiting M1 phenotype polarization and promoting microglia polarization toward the M2 phenotype, and its regulation of microglia M1/M2 polarization may be associated with modulating the BDNF/TrkB/CREB and Notch/Hes1 pathways.
Collapse
Affiliation(s)
- Yingyu Zhang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Danyang Wang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
- College of Food Science and Engineering, Shanxi Agricultural University, Jinzhong 030801, China
| | - Jiameng Liu
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Jing Sun
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Xinmin Liu
- Institute of New Drug Technology, Ningbo University, Ningbo 315211, China
| | - Bei Fan
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Cong Lu
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
- College of Food Science and Engineering, Shanxi Agricultural University, Jinzhong 030801, China
| | - Fengzhong Wang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
- College of Food Science and Engineering, Shanxi Agricultural University, Jinzhong 030801, China
| |
Collapse
|
39
|
Hu W, Zou L, Yu N, Wu Z, Yang W, Wu T, Liu Y, Pu Y, Jiang Y, Zhang J, Zhu H, Cheng F, Feng S. Catalpol rescues LPS-induced cognitive impairment via inhibition of NF-Κb-regulated neuroinflammation and up-regulation of TrkB-mediated BDNF secretion in mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117345. [PMID: 37926114 DOI: 10.1016/j.jep.2023.117345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/18/2023] [Accepted: 10/22/2023] [Indexed: 11/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Septic-associated encephalopathy (SAE) is a key manifestation of sepsis. Nevertheless, specific treatment for SAE is still lacking. Catalpol is an active component derived from Rehmanniae Radix, and has been demonstrated to be a potential neuroprotective agent. However, its effect on SAE still needs to be fully explored. AIM To address the benefits of catalpol on post-sepsis cognitive deterioration and related mechanisms. MATERIALS AND METHODS Novel object recognition test, temporal order task, histopathology, and immunochemistry were applied to address the benefits of catalpol on LPS-triggered post-sepsis cognitive decline in mice. Xuebijing injection (10 ml/kg) has been utilized as a positive control in the above animal studies. After treatment, the catalpol content in the hippocampus was determined using LC-MS/MS. Finally, the mechanisms of catalpol were further assessed in BV2 and PC12 cells in vitro using Western blot, RT-PCR, flow cytometry, molecular docking tests, thermal shift assay, transmission electron microscopy, and immunofluorescence analysis. RESULTS Behavior tests showed that catalpol therapy could lessen the cognitive impairment induced by LPS damage. HE, Nissl, immunofluorescence, transmission electron microscopy, and Golgi staining further reflected that catalpol treatment could restore lymphocyte infiltration, blood-brain barrier (BBB) degradation, and the decreasing complexity of dendritic trees. According to LC-MS/MS analysis, catalpol had a 136 ng/mg concentration in the hippocampus. In vitro investigation showed that catalpol could inhibit microglia M1 polarization via blocking NF-κB phosphorylation, translocation and then reducing inflammatory cytokine release in BV2 microglia cells. Brain-derived neurotrophic factor (BDNF) release up-regulation and TrkB pathway activation were observed in the catalpol treatment group in vivo and in vitro. The effect of catalpol on enhancing BDNF expression was inhibited by the specific inhibitor of TrkB (GNF-5837) in PC12 cells. Further molecular docking tests showed that catalpol formed weak hydrophobic bonds with TrkB. Besides, thermal shift assay also reflected that catalpol incubation caused a considerable change in the melting temperature of the TrkB. CONCLUSION Catalpol alleviates LPS-triggered post-sepsis cognitive impairment by reversing neuroinflammation via blocking the NF-κB pathway, up-regulating neurotrophic factors via the activation of TrkB pathway, and preserving BBB integrity.
Collapse
Affiliation(s)
- Weiqing Hu
- Department of Traditional Chinese Medicine, College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing, 400715, China.
| | - Li Zou
- Sichuan Vocational College of Health and Rehabilitation, Zigong, 643000, China.
| | - Ningxi Yu
- Department of Traditional Chinese Medicine, College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing, 400715, China.
| | - Zhizhongbin Wu
- Department of Traditional Chinese Medicine, College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing, 400715, China.
| | - Wei Yang
- Department of Traditional Chinese Medicine, College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing, 400715, China.
| | - Tianyue Wu
- Department of Traditional Chinese Medicine, College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing, 400715, China.
| | - Yulin Liu
- Department of Traditional Chinese Medicine, College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing, 400715, China.
| | - Yu Pu
- Department of Traditional Chinese Medicine, College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing, 400715, China.
| | - Yunbing Jiang
- Department of Traditional Chinese Medicine, College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing, 400715, China.
| | - Jifeng Zhang
- Department of Traditional Chinese Medicine, College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing, 400715, China
| | - Huifeng Zhu
- Department of Traditional Chinese Medicine, College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing, 400715, China.
| | - Fang Cheng
- The Third Affiliated Hospital of Henan University of Chinese Medicine, 63 Dongming Road, Zhengzhou City, Henan Province, China.
| | - Shan Feng
- Department of Traditional Chinese Medicine, College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
40
|
Avarlaid A, Esvald E, Koppel I, Parkman A, Zhuravskaya A, Makeyev EV, Tuvikene J, Timmusk T. An 840 kb distant upstream enhancer is a crucial regulator of catecholamine-dependent expression of the Bdnf gene in astrocytes. Glia 2024; 72:90-110. [PMID: 37632136 PMCID: PMC10952894 DOI: 10.1002/glia.24463] [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: 10/12/2022] [Revised: 08/10/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) plays a fundamental role in the developing and adult nervous system, contributing to neuronal survival, differentiation, and synaptic plasticity. Dysregulation of BDNF synthesis, secretion or signaling has been associated with many neurodevelopmental, neuropsychiatric, and neurodegenerative disorders. Although the transcriptional regulation of the Bdnf gene has been extensively studied in neurons, less is known about the regulation and function of BDNF in non-neuronal cells. The most abundant type of non-neuronal cells in the brain, astrocytes, express BDNF in response to catecholamines. However, genetic elements responsible for this regulation have not been identified. Here, we investigated four potential Bdnf enhancer regions and based on reporter gene assays, CRISPR/Cas9 engineering and CAPTURE-3C-sequencing we conclude that a region 840 kb upstream of the Bdnf gene regulates catecholamine-dependent expression of Bdnf in rodent astrocytes. We also provide evidence that this regulation is mediated by CREB and AP1 family transcription factors. This is the first report of an enhancer coordinating the transcription of Bdnf gene in non-neuronal cells.
Collapse
Affiliation(s)
- Annela Avarlaid
- Department of Chemistry and BiotechnologyTallinn University of TechnologyTallinnEstonia
| | - Eli‐Eelika Esvald
- Department of Chemistry and BiotechnologyTallinn University of TechnologyTallinnEstonia
- Protobios LLCTallinnEstonia
| | - Indrek Koppel
- Department of Chemistry and BiotechnologyTallinn University of TechnologyTallinnEstonia
| | - Annabel Parkman
- Department of Chemistry and BiotechnologyTallinn University of TechnologyTallinnEstonia
| | - Anna Zhuravskaya
- Centre for Developmental NeurobiologyKing's College LondonLondonUK
| | | | - Jürgen Tuvikene
- Department of Chemistry and BiotechnologyTallinn University of TechnologyTallinnEstonia
- Protobios LLCTallinnEstonia
| | - Tõnis Timmusk
- Department of Chemistry and BiotechnologyTallinn University of TechnologyTallinnEstonia
- Protobios LLCTallinnEstonia
| |
Collapse
|
41
|
Zhang XO, Zhang Y, Cho CE, Engelke DS, Smolen P, Byrne JH, Do-Monte FH. Enhancing Associative Learning in Rats With a Computationally Designed Training Protocol. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:165-181. [PMID: 38298784 PMCID: PMC10829654 DOI: 10.1016/j.bpsgos.2023.07.006] [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: 06/02/2023] [Revised: 06/30/2023] [Accepted: 07/20/2023] [Indexed: 02/02/2024] Open
Abstract
Background Learning requires the activation of protein kinases with distinct temporal dynamics. In Aplysia, nonassociative learning can be enhanced by a computationally designed learning protocol with intertrial intervals (ITIs) that maximize the interaction between fast-activated PKA (protein kinase A) and slow-activated ERK (extracellular signal-regulated kinase). Whether a similar strategy can enhance associative learning in mammals is unknown. Methods We simulated 1000 training protocols with varying ITIs to predict an optimal protocol based on empirical data for PKA and ERK dynamics in rat hippocampus. Adult male rats received the optimal protocol or control protocols in auditory fear conditioning and fear extinction experiments. Immunohistochemistry was performed to evaluate pCREB (phosphorylated cAMP response element binding)\protein levels in brain regions that have been implicated in fear acquisition. Results Rats exposed to the optimal conditioning protocol with irregular ITIs exhibited impaired extinction memory acquisition within the session using a standard footshock intensity, and stronger fear memory retrieval and spontaneous recovery with a weaker footshock intensity, compared with rats that received massed or spaced conditioning protocols with fixed ITIs. Rats exposed to the optimal extinction protocol displayed improved extinction of contextual fear memory and reduced spontaneous recovery compared with rats that received standard extinction protocols. Moreover, the optimal conditioning protocol increased pCREB levels in the dentate gyrus of the dorsal hippocampus, suggesting enhanced induction of long-term potentiation. Conclusions These findings demonstrate that a computational model-driven behavioral intervention can enhance associative learning in mammals and may provide insight into strategies to improve cognition in humans.
Collapse
Affiliation(s)
- Xu O. Zhang
- Department of Neurobiology and Anatomy, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Yili Zhang
- Department of Neurobiology and Anatomy, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Claire E. Cho
- Department of Neurobiology and Anatomy, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Douglas S. Engelke
- Department of Neurobiology and Anatomy, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Paul Smolen
- Department of Neurobiology and Anatomy, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas
| | - John H. Byrne
- Department of Neurobiology and Anatomy, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Fabricio H. Do-Monte
- Department of Neurobiology and Anatomy, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|
42
|
Hidalgo C, Paula-Lima A. RyR-mediated calcium release in hippocampal health and disease. Trends Mol Med 2024; 30:25-36. [PMID: 37957056 DOI: 10.1016/j.molmed.2023.10.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/16/2023] [Accepted: 10/20/2023] [Indexed: 11/15/2023]
Abstract
Hippocampal synaptic plasticity is widely considered the cellular basis of learning and spatial memory processes. This article highlights the central role of Ca2+ release from the endoplasmic reticulum (ER) in hippocampal synaptic plasticity and hippocampus-dependent memory in health and disease. The key participation of ryanodine receptor (RyR) channels, which are the principal Ca2+ release channels expressed in the hippocampus, in these processes is emphasized. It is proposed that the increased neuronal oxidative tone displayed by hippocampal neurons during aging or Alzheimer's disease (AD) leads to excessive activation of RyR-mediated Ca2+ release, a process that is highly redox-sensitive, and that this abnormal response contributes to and aggravates these deleterious conditions.
Collapse
Affiliation(s)
- Cecilia Hidalgo
- Biomedical Neuroscience Institute and Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile; Physiology and Biophysics Program, Institute of Biomedical Sciences and Center for Exercise, Metabolism, and Cancer Studies, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile.
| | - Andrea Paula-Lima
- Biomedical Neuroscience Institute and Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile; Institute for Research in Dental Sciences (ICOD), Faculty of Dentistry, Universidad de Chile, Santiago 8380544, Chile.
| |
Collapse
|
43
|
Sponchiado M, Bonilla AL, Mata L, Jasso-Johnson K, Liao YSJ, Fagan A, Moncada V, Reznikov LR. Club cell CREB regulates the goblet cell transcriptional network and pro-mucin effects of IL-1B. Front Physiol 2023; 14:1323865. [PMID: 38173934 PMCID: PMC10761479 DOI: 10.3389/fphys.2023.1323865] [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: 10/18/2023] [Accepted: 11/30/2023] [Indexed: 01/05/2024] Open
Abstract
Introduction: Club cells are precursors for mucus-producing goblet cells. Interleukin 1β (IL-1B) is an inflammatory mediator with pro-mucin activities that increases the number of mucus-producing goblet cells. IL-1B-mediated mucin production in alveolar adenocarcinoma cells requires activation of the cAMP response element-binding protein (CREB). Whether the pro-mucin activities of IL-1B require club cell CREB is unknown. Methods: We challenged male mice with conditional loss of club cell Creb1 and wild type littermates with intra-airway IL-1B or vehicle. Secondarily, we studied human "club cell-like" H322 cells. Results: IL-1B increased whole lung mRNA of secreted (Mucin 5ac, Mucin 5b) and tethered (Mucin 1, Mucin 4) mucins independent of genotype. However, loss of club cell Creb1 increased whole lung mRNA of member RAS oncogene family (Rab3D), decreased mRNA of the muscarinic receptor 3 (M3R) and prevented IL-1B mediated increases in purinergic receptor P2Y, (P2ry2) mRNA. IL-1B increased the density of goblet cells containing neutral mucins in wildtype mice but not in mice with loss of club cell Creb1. These findings suggested that club cell Creb1 regulated mucin secretion. Loss of club cell Creb1 also prevented IL-1B-mediated impairments in airway mechanics. Four days of pharmacologic CREB inhibition in H322 cells increased mRNA abundance of forkhead box A2 (FOXA2), a repressor of goblet cell expansion, and decreased mRNA expression of SAM pointed domain containing ETS transcription factor (SPDEF), a driver of goblet cell expansion. Chromatin immunoprecipitation demonstrated that CREB directly bound to the promoter region of FOXA2, but not to the promoter region of SPDEF. Treatment of H322 cells with IL-1B increased cAMP levels, providing a direct link between IL-1B and CREB signaling. Conclusion: Our findings suggest that club cell Creb1 regulates the pro-mucin properties of IL-1B through pathways likely involving FOXA2.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Leah R. Reznikov
- Department of Physiological Sciences, University of Florida, Gainesville, FL, United States
| |
Collapse
|
44
|
Cheng H, Villahoz BF, Ponzio RD, Aschner M, Chen P. Signaling Pathways Involved in Manganese-Induced Neurotoxicity. Cells 2023; 12:2842. [PMID: 38132161 PMCID: PMC10742340 DOI: 10.3390/cells12242842] [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: 11/01/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
Manganese (Mn) is an essential trace element, but insufficient or excessive bodily amounts can induce neurotoxicity. Mn can directly increase neuronal insulin and activate insulin-like growth factor (IGF) receptors. As an important cofactor, Mn regulates signaling pathways involved in various enzymes. The IGF signaling pathway plays a protective role in the neurotoxicity of Mn, reducing apoptosis in neurons and motor deficits by regulating its downstream protein kinase B (Akt), mitogen-activated protein kinase (MAPK), and mammalian target of rapamycin (mTOR). In recent years, some new mechanisms related to neuroinflammation have been shown to also play an important role in Mn-induced neurotoxicity. For example, DNA-sensing receptor cyclic GMP-AMP synthase (cCAS) and its downstream signal efficient interferon gene stimulator (STING), NOD-like receptor family pyrin domain containing 3(NLRP3)-pro-caspase1, cleaves to the active form capase1 (CASP1), nuclear factor κB (NF-κB), sirtuin (SIRT), and Janus kinase (JAK) and signal transducers and activators of the transcription (STAT) signaling pathway. Moreover, autophagy, as an important downstream protein degradation pathway, determines the fate of neurons and is regulated by these upstream signals. Interestingly, the role of autophagy in Mn-induced neurotoxicity is bidirectional. This review summarizes the molecular signaling pathways of Mn-induced neurotoxicity, providing insight for further understanding of the mechanisms of Mn.
Collapse
Affiliation(s)
| | | | | | | | - Pan Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (H.C.); (B.F.V.); (R.D.P.); (M.A.)
| |
Collapse
|
45
|
Hamad MIK, Emerald BS, Kumar KK, Ibrahim MF, Ali BR, Bataineh MF. Extracellular molecular signals shaping dendrite architecture during brain development. Front Cell Dev Biol 2023; 11:1254589. [PMID: 38155836 PMCID: PMC10754048 DOI: 10.3389/fcell.2023.1254589] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/28/2023] [Indexed: 12/30/2023] Open
Abstract
Proper growth and branching of dendrites are crucial for adequate central nervous system (CNS) functioning. The neuronal dendritic geometry determines the mode and quality of information processing. Any defects in dendrite development will disrupt neuronal circuit formation, affecting brain function. Besides cell-intrinsic programmes, extrinsic factors regulate various aspects of dendritic development. Among these extrinsic factors are extracellular molecular signals which can shape the dendrite architecture during early development. This review will focus on extrinsic factors regulating dendritic growth during early neuronal development, including neurotransmitters, neurotrophins, extracellular matrix proteins, contact-mediated ligands, and secreted and diffusible cues. How these extracellular molecular signals contribute to dendritic growth has been investigated in developing nervous systems using different species, different areas within the CNS, and different neuronal types. The response of the dendritic tree to these extracellular molecular signals can result in growth-promoting or growth-limiting effects, and it depends on the receptor subtype, receptor quantity, receptor efficiency, the animal model used, the developmental time windows, and finally, the targeted signal cascade. This article reviews our current understanding of the role of various extracellular signals in the establishment of the architecture of the dendrites.
Collapse
Affiliation(s)
- Mohammad I. K. Hamad
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Kukkala K. Kumar
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Marwa F. Ibrahim
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Bassam R. Ali
- Department of Genetics and Genomics, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mo’ath F. Bataineh
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
46
|
Lee CH, Ko MS, Kim YS, Ham JE, Choi JY, Hwang KW, Park SY. Neuroprotective Effects of Davallia mariesii Roots and Its Active Constituents on Scopolamine-Induced Memory Impairment in In Vivo and In Vitro Studies. Pharmaceuticals (Basel) 2023; 16:1606. [PMID: 38004471 PMCID: PMC10675602 DOI: 10.3390/ph16111606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Beta-amyloid (Aβ) proteins, major contributors to Alzheimer's disease (AD), are overproduced and accumulate as oligomers and fibrils. These protein accumulations lead to significant changes in neuronal structure and function, ultimately resulting in the neuronal cell death observed in AD. Consequently, substances that can inhibit Aβ production and/or accumulation are of great interest for AD prevention and treatment. In the course of an ongoing search for natural products, the roots of Davallia mariesii T. Moore ex Baker were selected as a promising candidate with anti-amyloidogenic effects. The ethanol extract of D. mariesii roots, along with its active constituents, not only markedly reduced Aβ production by decreasing β-secretase expression in APP-CHO cells (Chinese hamster ovary cells which stably express amyloid precursor proteins), but also exhibited the ability to diminish Aβ aggregation while enhancing the disaggregation of Aβ aggregates, as determined through the Thioflavin T (Th T) assay. Furthermore, in an in vivo study, the extract of D. mariesii roots showed potential (a tendency) for mitigating scopolamine-induced memory impairment, as evidenced by results from the Morris water maze test and the passive avoidance test, which correlated with reduced Aβ deposition. Additionally, the levels of acetylcholine were significantly elevated, and acetylcholinesterase levels significantly decreased in the brains of mice (whole brains). The treatment with the extract of D. mariesii roots also led to upregulated brain-derived neurotrophic factor (BDNF) and phospho-cAMP response element-binding protein (p-CREB) in the hippocampal region. These findings suggest that the extract of D. mariesii roots, along with its active constituents, may offer neuroprotective effects against AD. Consequently, there is potential for the development of the extract of D. mariesii roots and its active constituents as effective therapeutic or preventative agents for AD.
Collapse
Affiliation(s)
- Chung Hyeon Lee
- College of Pharmacy, Dankook University, Cheonan 31116, Republic of Korea; (C.H.L.); (M.S.K.); (Y.S.K.)
| | - Min Sung Ko
- College of Pharmacy, Dankook University, Cheonan 31116, Republic of Korea; (C.H.L.); (M.S.K.); (Y.S.K.)
| | - Ye Seul Kim
- College of Pharmacy, Dankook University, Cheonan 31116, Republic of Korea; (C.H.L.); (M.S.K.); (Y.S.K.)
| | - Ju Eon Ham
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea; (J.E.H.); (J.Y.C.)
| | - Jee Yeon Choi
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea; (J.E.H.); (J.Y.C.)
| | - Kwang Woo Hwang
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea; (J.E.H.); (J.Y.C.)
| | - So-Young Park
- College of Pharmacy, Dankook University, Cheonan 31116, Republic of Korea; (C.H.L.); (M.S.K.); (Y.S.K.)
| |
Collapse
|
47
|
Zernov N, Popugaeva E. Role of Neuronal TRPC6 Channels in Synapse Development, Memory Formation and Animal Behavior. Int J Mol Sci 2023; 24:15415. [PMID: 37895105 PMCID: PMC10607207 DOI: 10.3390/ijms242015415] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
The transient receptor potential cation channel, subfamily C, member 6 (TRPC6), has been believed to adjust the formation of an excitatory synapse. The positive regulation of TRPC6 engenders synapse enlargement and improved learning and memory in animal models. TRPC6 is involved in different synaptoprotective signaling pathways, including antagonism of N-methyl-D-aspartate receptor (NMDAR), activation of brain-derived neurotrophic factor (BDNF) and postsynaptic store-operated calcium entry. Positive regulation of TRPC6 channels has been repeatedly shown to be good for memory formation and storage. TRPC6 is mainly expressed in the hippocampus, particularly in the dentate granule cells, cornu Ammonis 3 (CA3) pyramidal cells and gamma-aminobutyric acid (GABA)ergic interneurons. It has been observed that TRPC6 agonists have a great influence on animal behavior including memory formation and storage The purpose of this review is to collect the available information on the role of TRPC6 in memory formation in various parts of the brain to understand how TRPC6-specific pharmaceutical agents will affect memory in distinct parts of the central nervous system (CNS).
Collapse
Affiliation(s)
| | - Elena Popugaeva
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia
| |
Collapse
|
48
|
Cefis M, Chaney R, Wirtz J, Méloux A, Quirié A, Leger C, Prigent-Tessier A, Garnier P. Molecular mechanisms underlying physical exercise-induced brain BDNF overproduction. Front Mol Neurosci 2023; 16:1275924. [PMID: 37868812 PMCID: PMC10585026 DOI: 10.3389/fnmol.2023.1275924] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
Accumulating evidence supports that physical exercise (EX) is the most effective non-pharmacological strategy to improve brain health. EX prevents cognitive decline associated with age and decreases the risk of developing neurodegenerative diseases and psychiatric disorders. These positive effects of EX can be attributed to an increase in neurogenesis and neuroplastic processes, leading to learning and memory improvement. At the molecular level, there is a solid consensus to involve the neurotrophin brain-derived neurotrophic factor (BDNF) as the crucial molecule for positive EX effects on the brain. However, even though EX incontestably leads to beneficial processes through BDNF expression, cellular sources and molecular mechanisms underlying EX-induced cerebral BDNF overproduction are still being elucidated. In this context, the present review offers a summary of the different molecular mechanisms involved in brain's response to EX, with a specific focus on BDNF. It aims to provide a cohesive overview of the three main mechanisms leading to EX-induced brain BDNF production: the neuronal-dependent overexpression, the elevation of cerebral blood flow (hemodynamic hypothesis), and the exerkine signaling emanating from peripheral tissues (humoral response). By shedding light on these intricate pathways, this review seeks to contribute to the ongoing elucidation of the relationship between EX and cerebral BDNF expression, offering valuable insights into the potential therapeutic implications for brain health enhancement.
Collapse
Affiliation(s)
- Marina Cefis
- Département des Sciences de l’Activité Physique, Faculté des Sciences, Université du Québec à Montréal, Montreal, QC, Canada
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Remi Chaney
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Julien Wirtz
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Alexandre Méloux
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Aurore Quirié
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Clémence Leger
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Anne Prigent-Tessier
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Philippe Garnier
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
- Département Génie Biologique, Institut Universitaire de Technologie, Dijon, France
| |
Collapse
|
49
|
Anyachor CP, Orish CN, Ezejiofor AN, Cirovic A, Cirovic A, Ezealisiji KM, Orisakwe OE. Nickel and aluminium mixture elicit memory impairment by activation of oxidative stress, COX-2, and diminution of AChE, BDNF and NGF levels in cerebral cortex and hippocampus of male albino rats. Curr Res Toxicol 2023; 5:100129. [PMID: 37841055 PMCID: PMC10569962 DOI: 10.1016/j.crtox.2023.100129] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 10/17/2023] Open
Abstract
This study evaluated nickel and aluminium-induced neurotoxicity, as a binary metal mixture. Twenty-eight male Sprague Dawley albino rats were weight-matched and divided into four groups. Group 1 (control) received deionized water. Group 2 and 3 received Aluminium (1 mg/kg) and Nickel (0.2 mg/kg) respectively, while Group 4 received Ni and Al mixture HMM three times a week orally for 90 days. Barnes maze tests was performed. Rats were sacrificed under pentobarbital anaesthesia, cerebral cortex and hippocampus were separated, and metal levels were measured using Atomic Absorption Spectroscopy (AAS). Malondialdehyde (MDA), catalase (CAT), glutathione content (GSH), superoxide dismutase (SOD), glutathione peroxidase (GPx), Brain Derived Neurotrophic Factor (BDNF), Nerve growth factor NGF, cyclo-oxygenase COX-2 and Acetylcholinesterase (AChE) were assayed using ELISA kits. Ni/Al binary mixture exposed rats showed a shorter latency period (though not significant) of 3.21 ± 1.40 s in comparison to 3.77 ± 1.11 (Ni only) and 3.99 ± 1.16(Al only). Ni/Al mixture gp had the lowest levels of Mg in both the hippocampus and frontal cortex when compared with the individual metals. In the hippocampus Al only exposed rats significantly showed p < 0.05 higher iron and Ca levels in comparison to Ni/Al mixture. Al alone significantly showed p < 0.05 lower levels of Fe but higher Ca than the Ni/Al mixture group. Exposure to Al only showed lower levels of BDNF in comparison to Ni/Al combination, whereas Ni/Al mixture gp had lower levels of NGF in comparison to the individual metals in the hippocampus. In the frontal cortex Ni only, group showed significantly lower BDNF in comparison to Ni/Al mixture whereas the mixture showed significantly lower NGF when compared with Al only group. There were higher levels of COX-2 in the Ni/Al mixture than individual metal treated rats in both hippocampus and frontal cortex. AChE levels in the Ni/Al mixture group was higher than Ni or Al only gps in the hippocampus whereas in the frontal cortex, Ni/Al exposed rats showed significantly lower AChE levels in comparison to Al only group. Ni, Al and Ni/Al mixture exhibited memory impairment by activation of oxidative stress, COX-2, and diminution of AChE, BDNF and NGF levels in cerebral cortex and hippocampus. The BDNF-COX-2 AChE signalling pathway may be involved in the neurotoxicity of Ni and Al.
Collapse
Affiliation(s)
- Chidinma P. Anyachor
- African Centre of Excellence for Public Health and Toxicological Research (ACE-PUTOR), University of Port Harcourt, PMB, 5323, Choba, Port Harcourt, Nigeria
| | - Chinna N. Orish
- African Centre of Excellence for Public Health and Toxicological Research (ACE-PUTOR), University of Port Harcourt, PMB, 5323, Choba, Port Harcourt, Nigeria
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Port Harcourt, PMB, 5323, Choba, Port Harcourt, Nigeria
| | - Anthonet N. Ezejiofor
- African Centre of Excellence for Public Health and Toxicological Research (ACE-PUTOR), University of Port Harcourt, PMB, 5323, Choba, Port Harcourt, Nigeria
| | - Ana Cirovic
- University of Belgrade, Faculty of Medicine, Institute of Anatomy, Belgrade, Serbia
| | - Aleksandar Cirovic
- University of Belgrade, Faculty of Medicine, Institute of Anatomy, Belgrade, Serbia
| | - Kenneth M. Ezealisiji
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Port Harcourt, PMB, 5323, Choba, Port Harcourt, Nigeria
| | - Orish E. Orisakwe
- African Centre of Excellence for Public Health and Toxicological Research (ACE-PUTOR), University of Port Harcourt, PMB, 5323, Choba, Port Harcourt, Nigeria
| |
Collapse
|
50
|
Thapak P, Smith G, Ying Z, Paydar A, Harris N, Gomez-Pinilla F. The BDNF mimetic R-13 attenuates TBI pathogenesis using TrkB-related pathways and bioenergetics. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166781. [PMID: 37286142 PMCID: PMC10619508 DOI: 10.1016/j.bbadis.2023.166781] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/22/2023] [Accepted: 05/30/2023] [Indexed: 06/09/2023]
Abstract
Traumatic brain injury (TBI) is major neurological burden globally, and effective treatments are urgently needed. TBI is characterized by a reduction in energy metabolism and synaptic function that seems a primary cause of neuronal dysfunction. R13, a small drug and BDNF mimetic showed promising results in improving spatial memory and anxiety-like behavior after TBI. Additionally, R13 was found to counteract reductions in molecules associated with BDNF signaling (p-TrkB, p-PI3K, p-AKT), synaptic plasticity (GluR2, PSD95, Synapsin I) as well as bioenergetic components such as mitophagy (SOD, PGC-1α, PINK1, Parkin, BNIP3, and LC3) and real-time mitochondrial respiratory capacity. Behavioral and molecular changes were accompanied by adaptations in functional connectivity assessed using MRI. Results highlight the potential of R13 as a therapeutic agent for TBI and provide valuable insights into the molecular and functional changes associated with this condition.
Collapse
Affiliation(s)
- Pavan Thapak
- Dept. Integrative Biology and Physiology, UCLA, Los Angeles, CA, United States of America
| | - Gregory Smith
- Department of Neurosurgery, UCLA David Geffen School of Medicine, Los Angeles, CA, United States of America; UCLA Brain Injury Research Center, Los Angeles, CA, United States of America
| | - Zhe Ying
- Dept. Integrative Biology and Physiology, UCLA, Los Angeles, CA, United States of America
| | - Afshin Paydar
- Department of Neurosurgery, UCLA David Geffen School of Medicine, Los Angeles, CA, United States of America; UCLA Brain Injury Research Center, Los Angeles, CA, United States of America
| | - Neil Harris
- Department of Neurosurgery, UCLA David Geffen School of Medicine, Los Angeles, CA, United States of America; UCLA Brain Injury Research Center, Los Angeles, CA, United States of America; Intellectual Development and Disabilities Research Center, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Fernando Gomez-Pinilla
- Dept. Integrative Biology and Physiology, UCLA, Los Angeles, CA, United States of America; Department of Neurosurgery, UCLA David Geffen School of Medicine, Los Angeles, CA, United States of America; UCLA Brain Injury Research Center, Los Angeles, CA, United States of America.
| |
Collapse
|