1
|
Shalaby HN, Nawwar DA, Kamel AS, El-Said YA. Blocking hippocampal voltage-gated potassium channel Kv1.3 by dalfampridine abrogates cognitive impairment in experimental autoimmune encephalomyelitis mouse model. Eur J Pharmacol 2025; 998:177647. [PMID: 40252897 DOI: 10.1016/j.ejphar.2025.177647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 03/25/2025] [Accepted: 04/17/2025] [Indexed: 04/21/2025]
Abstract
Multiple sclerosis (MS) is devastating motor disorders accompanied by cognitive impairments. Dalfampridine (Dal), was approved for treating MS via blocking voltage-gated potassium channel (Kv). Kv1.3 is one of Dals' targets that showed insults to cognitive function in preclinical and clinical cases. Yet, there is no study that has assessed the potentiality of Dal on Kv1.3-induced cognitive impairment in MS. Thus, the aim of the present study is to reveal the procognitive influence of Dal in the MS animal model. Mice were randomly assigned into four groups. Saline was administered to group 1, conversely groups 2, 3, and 4 received 2 shots of rat spinal cord emulsified with complete Freund's adjuvant. Group 3 received Dal (10 μg/mouse; p. o), while group 4 received Wortmannin (0.1 μg/mouse; i. c.v), a selective phosphoinositide 3-kinases (PI3K) inhibitor, before Dal administration. After 14 days, Dal alleviated motor deficits in the open field arena and rotarod with cognitive restoration in the novel object recognition task. These were accompanied by the reversal of hippocampal neuronal loss and demyelination in corpus callosum. Additionally, Dal inhibited Kv1.3 that enhanced survival signaling, viz; PI3K/Akt. Such activation abates neuroinflammatory markers, glycogen synthase kinase-3 beta (GSK-3β) and nuclear factor-κB levels with subsequent enhancement of BDNF. All these amendments were reversed by Wortmannin pre-administration. In conclusion, this study declares that blockade of Kv1.3 and modulation of PI3K/Akt/GSK-3β/BDNF axis with Dal could be proposed as a promising neuroprotective and memory-enhancing treatment in MS.
Collapse
MESH Headings
- Animals
- Kv1.3 Potassium Channel/antagonists & inhibitors
- Kv1.3 Potassium Channel/metabolism
- Hippocampus/drug effects
- Hippocampus/metabolism
- Hippocampus/pathology
- Cognitive Dysfunction/drug therapy
- Cognitive Dysfunction/metabolism
- Cognitive Dysfunction/etiology
- Mice
- Potassium Channel Blockers/pharmacology
- Potassium Channel Blockers/therapeutic use
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/complications
- Encephalomyelitis, Autoimmune, Experimental/psychology
- 4-Aminopyridine/pharmacology
- 4-Aminopyridine/therapeutic use
- 4-Aminopyridine/analogs & derivatives
- Disease Models, Animal
- Rats
- Male
- Brain-Derived Neurotrophic Factor/metabolism
- Female
- Behavior, Animal/drug effects
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Heba Nasr Shalaby
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Dalia A Nawwar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ahmed S Kamel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, Egypt
| | - Yasmin Am El-Said
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
2
|
Laabi S, LeMmon C, Vogel C, Chacon M, Jimenez VM. Psilocybin and psilocin regulate microglial immunomodulation and support neuroplasticity via serotonergic and AhR signaling. Int Immunopharmacol 2025; 159:114940. [PMID: 40424654 DOI: 10.1016/j.intimp.2025.114940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/22/2025] [Accepted: 05/20/2025] [Indexed: 05/29/2025]
Abstract
BACKGROUND Psilocybin, a serotonergic psychedelic, has demonstrated therapeutic potential in neuropsychiatric disorders. While its neuroplastic and immunomodulatory effects are recognized, the underlying mechanisms remain unclear. This study investigates how psilocybin and its active metabolite, psilocin, influence microglial inflammatory responses and neurotrophic factor expression through serotonergic and AhR signaling. METHODS Using in vitro models of resting and LPS-activated microglia, we evaluated the effects of psilocybin and psilocin on the expression of pro-inflammatory cytokines (TNF-α), anti-inflammatory cytokines (IL-10), and neuroplasticity-related markers (BDNF). Receptor-specific contributions were assessed using selective antagonists for 5-HT2A, 5-HT2B, 5-HT7, TrkB, and AhR. RESULTS Psilocybin and psilocin significantly suppressed TNF-α expression and increased BDNF levels in LPS-activated microglia. These effects were mediated by 5-HT2A, 5-HT2B, 5-HT7, and TrkB signaling, while AhR activation was required for psilocin-induced BDNF upregulation but not TNF-α suppression. IL-10 levels remained unchanged under normal conditions but increased significantly when serotonergic, TrkB, or AhR signaling was blocked, suggesting a compensatory shift in anti-inflammatory pathways. CONCLUSION Psilocybin and psilocin promote a microglial phenotype that reduces inflammation and supports neuroplasticity via receptor-specific mechanisms. Their effects on TNF-α and BDNF depend on distinct serotonergic and neurotrophic pathways, with AhR playing a selective role in psilocin's action. These findings clarify the receptor-mediated dynamics of psilocybin's therapeutic effects and highlight alternative anti-inflammatory pathways that may be relevant for clinical applications.
Collapse
Affiliation(s)
- Salma Laabi
- Department of Biomedical Sciences, Noorda College of Osteopathic Medicine, 2162 S 180 E, Provo, UT 84606, United States
| | - Claire LeMmon
- Department of Biomedical Sciences, Noorda College of Osteopathic Medicine, 2162 S 180 E, Provo, UT 84606, United States
| | - Callie Vogel
- Department of Biomedical Sciences, Noorda College of Osteopathic Medicine, 2162 S 180 E, Provo, UT 84606, United States
| | - Mariana Chacon
- Department of Biomedical Sciences, Noorda College of Osteopathic Medicine, 2162 S 180 E, Provo, UT 84606, United States
| | - Victor M Jimenez
- Department of Biomedical Sciences, Noorda College of Osteopathic Medicine, 2162 S 180 E, Provo, UT 84606, United States; Department of Pharmacy, Roseman University of Health Sciences, 10920 S River Front Pkwy, South Jordan, UT 84095, United States.
| |
Collapse
|
3
|
Lee NK, Lee Y, Won M, Kim N, Park E, Paik HD. Preventive Effect of Probiotic Leuconostoc mesenteroides H40 Against Cognitive Disorder by Anti-Inflammatory, Synaptic Plasticity Regulation, and Antioxidant Effects. Antioxidants (Basel) 2025; 14:565. [PMID: 40427447 DOI: 10.3390/antiox14050565] [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: 04/08/2025] [Revised: 05/01/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Live Leuconostoc mesenteroides H40 has been reported to have probiotic properties; however, the effect of its live probiotic form on cognitive ability has not been reported. This study investigated modulatory effects of the probiotic L. mesenteroides H40 in an ICR mouse model (male) of cognitive disorders. Cognitive disorders were induced in mice by the addition of scopolamine (1 mg/kg/day) with donepezil (2 mg/kg/day) as a medicinal control. L. mesenteroides H40 significantly attenuated scopolamine-induced cognitive disorder in the novel object recognition and Y-maze tests in a concentration-dependent manner. L. mesenteroides H40 decreased amyloid β levels, but increased β-secretase levels. The mRNA expression levels of inducible nitric oxide synthase (iNOS) and cyclooxygenase (COX)-2 significantly decreased following L. mesenteroides H40 treatment. Additionally, TNF-α, IL-1β, and PGE2 protein expression was decreased. Acetylcholine, acetylcholinesterase, choline acetyltransferase, brain-derived neurotrophic factor (BDNF), and cAMP response element-binding protein (CREB) levels were increased in the brain tissues. The antioxidant effects of superoxide dismutase, catalase, and glutathione peroxidase were also alleviated. We demonstrated that L. mesenteroides H40 exhibits neuroprotective effects through anti-inflammatory, synaptic plasticity regulation, and antioxidant effects. Thus, the probiotic L. mesenteroides H40 could be used as a prophylactic functional food for cognitive disorders.
Collapse
Affiliation(s)
- Na-Kyoung Lee
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Republic of Korea
| | - Yunjung Lee
- Department of Food and Nutrition, Kyungnam University, Changwon 51767, Republic of Korea
| | - Minhye Won
- Department of Food and Nutrition, Kyungnam University, Changwon 51767, Republic of Korea
| | - Nayeong Kim
- Department of Food and Nutrition, Kyungnam University, Changwon 51767, Republic of Korea
| | - Eunju Park
- Department of Food and Nutrition, Kyungnam University, Changwon 51767, Republic of Korea
| | - Hyun-Dong Paik
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Republic of Korea
- View of Creativity, GHBio Co., Ltd., 120 Neungdong-Ro, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
4
|
Sun Y, Wang J, Ding W, Qin Q, Wang R, Yu R, Yan J, Hou R, Liu G, Cai X, Qu Z, Zhang W, Yu J, Xing C, Zhuang C. Design of anti-depressant phosphodiester amino acidic Keap1-Nrf2 protein-protein interaction inhibitors. Redox Biol 2025; 82:103620. [PMID: 40174476 PMCID: PMC11999321 DOI: 10.1016/j.redox.2025.103620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025] Open
Abstract
Inhibiting the protein-protein interaction (PPI) between Keap1 and Nrf2 is theoretically an effective and safe strategy for activation of Nrf2 pathway to treat major depressive disorder (MDD). In this study, through bioinformatic analysis of the brain tissues and peripheral blood of MDD patients and depressive mice, we confirmed the involvement of oxidative stress, inflammation, and the Keap1-Nrf2 pathway in depression. Subsequently, we developed a series of phosphodiester amino acidic diaminonaphthalene compounds as Keap1-Nrf2 PPI inhibitors for the first time. Screening using the LPS-stimulated SH-SY5Y and BV2 cell models identified compound 4-95 showing the best anti-oxidative stress and anti-inflammatory efficacy. The ability of 4-95 to penetrate the blood-brain-barrier was significantly enhanced. In a chronic unpredictable mild stress mouse model, treatment with 4-95 effectively ameliorated anxiety and depression behavior and restored serum neurotransmitter levels by promoting the Nrf2 nuclear translocation. Consequently, oxidative stress was reduced, and the expression of synaptic plasticity biomarkers, such as postsynaptic density protein 95 (PSD95) and synapsin 1 (SYN1) were significantly increased, suggesting the recovery of neuronal function. Collectively, our findings demonstrate that the Keap1-Nrf2 PPI inhibitor holds great promise as a preclinical candidate for the treatment of depression.
Collapse
Affiliation(s)
- Yi Sun
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, 639 Longmian Dadao, Jiangning District, Nanjing, 210009, China
| | - Jue Wang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, 639 Longmian Dadao, Jiangning District, Nanjing, 210009, China
| | - Wenxin Ding
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| | - Qingqing Qin
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| | - Rui Wang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, 639 Longmian Dadao, Jiangning District, Nanjing, 210009, China
| | - Ruizhi Yu
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| | - Jianyu Yan
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| | - Ruilin Hou
- Department of Pharmacy, Drug/Medical Device Clinical Trial Institution Office, General Hospital of Ningxia Medical University, 804 Shengli Street, Yinchuan, Ningxia, 750003, China
| | - Guodong Liu
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China
| | - Xiangming Cai
- Department of Pharmacy, Nantong Fourth People's Hospital, 37 Chenggang Road, Nantong, 226000, China
| | - Zhuo Qu
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China
| | - Wannian Zhang
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China; School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China
| | - Jianqiang Yu
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China
| | - Chengguo Xing
- Department of Medicinal Chemistry, University of Florida, 1345 Center Drive, Gainesville, FL, 32610, USA
| | - Chunlin Zhuang
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China; School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China.
| |
Collapse
|
5
|
Guo Y, Li J, Liu X, Ding H, Zhang W. Potential therapeutic targets for ischemic stroke in pre-clinical studies: Epigenetic-modifying enzymes DNMT/TET and HAT/HDAC. Front Pharmacol 2025; 16:1571276. [PMID: 40356977 PMCID: PMC12066669 DOI: 10.3389/fphar.2025.1571276] [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: 02/05/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Ischemic stroke (IS) remains a leading cause of mortality and disability worldwide, driven by genetic predispositions and environmental interactions, with epigenetics playing a pivotal role in mediating these processes. Specific modifying enzymes that regulate epigenetic changes have emerged as promising targets for IS treatment. DNA methyltransferases (DNMTs), ten-eleven translocation (TET) dioxygenases, histone acetyltransferases (HATs), and histone deacetylases (HDACs) are central to epigenetic regulation. These enzymes maintain a dynamic balance between DNA methylation/demethylation and histone acetylation/deacetylation, which critically influences gene expression and neuronal survival in IS. This review is based on both in vivo and in vitro experimental studies, exploring the roles of DNMT/TET and HAT/HDAC in IS, evaluating their potential as therapeutic targets, and discussing the use of natural compounds as modulators of these enzymes to develop novel treatment strategies.
Collapse
Affiliation(s)
- Yurou Guo
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Jing Li
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Xiaodan Liu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
- Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Huang Ding
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
- Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Wei Zhang
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
- Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| |
Collapse
|
6
|
Torres VO, Turchan-Cholewo J, Colson MK, Yanev P, Britsch DRS, Cotter KM, McAtee AM, Ujas TA, Mercurio D, Kong X, Plautz EJ, Joshi CR, Matsui TK, Mori E, Cajigas-Hernandez A, Zuurbier K, Estus S, Goldberg MP, Monson NL, Stowe AM. B cells upregulate NMDARs, respond to extracellular glutamate, and express mature BDNF to protect the brain from ischemic injury. Neurobiol Dis 2025; 207:106819. [PMID: 39900302 PMCID: PMC11948303 DOI: 10.1016/j.nbd.2025.106819] [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: 10/16/2024] [Revised: 01/27/2025] [Accepted: 01/27/2025] [Indexed: 02/05/2025] Open
Abstract
Following stroke, B cells enter brain regions outside of the ischemic injury to mediate functional recovery. Although B cells produce neurotrophins that support remote plasticity, including brain-derived neurotrophic factor (BDNF), it remains unclear which signal(s) activate B cells in the absence of infarct-localized pro-inflammatory cues. Activation of N-methyl-d-aspartate (NMDA)-type receptor (NMDAR) subunits on neurons can upregulate mature BDNF (mBDNF) production from a pro-BDNF precursor, but whether this occurs in B cells is unknown. We identified GluN2A and GluN2B NMDAR subunits on B cells that respond to glutamate and mediate nearly half of the glutamate-induced Ca2+ responses in activated B cell subsets. Ischemic stroke recruits GluN2A+ B cells into the ipsilesional hemisphere and both stroke and neurophysiologic levels of glutamate regulate gene and surface expression. Regardless of injury, pro-BDNF+ B cells localize to spleen/circulation whereas mBDNF+ B cells localize to the brain, including in aged male and female mice. We confirmed B cell-derived BDNF was required for in vitro and in vivo B cell-mediated neuroprotection. Lastly, GluN2A, GluN2B, glutamate-induced Ca2+ responses, and BDNF expression were all clinically confirmed in B cells from healthy donors, with BDNF+ B cells present in post-stroke human parenchyma. These data suggest that B cells express functional NMDARs that respond to glutamate, enhance NMDAR signaling with activation, and upregulate mature BDNF expression within the brain. This study identifies potential glutamate-induced neurotrophic roles for B cells in the brain; an immune response to neurotransmitters unique from established pro-inflammatory stimuli and relevant to any CNS-localized injury or disease.
Collapse
Affiliation(s)
- Vanessa O Torres
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Suite NL9.114, Dallas, TX 75390-8813, USA; Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Jadwiga Turchan-Cholewo
- Department of Neurology, University of Kentucky, 740 S. Limestone, Kentucky Clinic J-455, Lexington, KY 40536, USA
| | - Mary K Colson
- Department of Neurology, University of Kentucky, 740 S. Limestone, Kentucky Clinic J-455, Lexington, KY 40536, USA
| | - Pavel Yanev
- Department of Neurology, University of Kentucky, 740 S. Limestone, Kentucky Clinic J-455, Lexington, KY 40536, USA
| | - Daimen R S Britsch
- Department of Neurology, University of Kentucky, 740 S. Limestone, Kentucky Clinic J-455, Lexington, KY 40536, USA
| | - Katherine M Cotter
- Department of Neurology, University of Kentucky, 740 S. Limestone, Kentucky Clinic J-455, Lexington, KY 40536, USA
| | - Annabel M McAtee
- Department of Neurology, University of Kentucky, 740 S. Limestone, Kentucky Clinic J-455, Lexington, KY 40536, USA
| | - Thomas A Ujas
- Department of Neurology, University of Kentucky, 740 S. Limestone, Kentucky Clinic J-455, Lexington, KY 40536, USA
| | - Domenico Mercurio
- Department of Neurology, University of Kentucky, 740 S. Limestone, Kentucky Clinic J-455, Lexington, KY 40536, USA
| | - Xiangmei Kong
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Suite NL9.114, Dallas, TX 75390-8813, USA
| | - Erik J Plautz
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Suite NL9.114, Dallas, TX 75390-8813, USA
| | - Chaitanya R Joshi
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Suite NL9.114, Dallas, TX 75390-8813, USA
| | - Takeshi K Matsui
- Department of Neural and Muscular Physiology, Shimane University School of Medicine, 89-1 Enya-cho, Izumo-shi 693-8501, Shimane, Japan; Department of Future Basic Medicine, Nara Medical University, 840 Shijo-Cho, Kashihara, 634-8521 Nara, Japan
| | - Eiichiro Mori
- Department of Future Basic Medicine, Nara Medical University, 840 Shijo-Cho, Kashihara, 634-8521 Nara, Japan
| | - Ambar Cajigas-Hernandez
- Department of Neuroscience, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9111, USA
| | - Kielen Zuurbier
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9148, USA
| | - Steven Estus
- Department of Physiology, University of Kentucky, 741 S. Limestone, BBSRB B243, Lexington, KY 40536, USA
| | - Mark P Goldberg
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Suite NL9.114, Dallas, TX 75390-8813, USA; Department of Neurology, Institute for Integration of Medicine and Science, UT Health San Antonio, 7703 Floyd Curl Drive, MSC 7883, San Antonio, TX 78229, USA
| | - Nancy L Monson
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Suite NL9.114, Dallas, TX 75390-8813, USA; Department of Immunology, University of Texas Southwestern Medical Center, 6124 Harry Hines Blvd., Dallas, TX 75390-9093, USA
| | - Ann M Stowe
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Suite NL9.114, Dallas, TX 75390-8813, USA; Department of Neurology, University of Kentucky, 740 S. Limestone, Kentucky Clinic J-455, Lexington, KY 40536, USA.
| |
Collapse
|
7
|
Hong J, Wu Y, Li M, Man KF, Song D, Koh SB. cAMP response element-binding protein: A credible cancer drug target. J Pharmacol Exp Ther 2025; 392:103529. [PMID: 40157009 PMCID: PMC12060161 DOI: 10.1016/j.jpet.2025.103529] [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: 08/17/2024] [Accepted: 02/22/2025] [Indexed: 04/01/2025] Open
Abstract
Despite advancements in radiotherapy, chemotherapy, endocrine therapy, targeted therapy, and immunotherapy, resistance to therapy remains a pervasive challenge in oncology, in part owing to tumor heterogeneity. Identifying new therapeutic targets is key to addressing this challenge because it can both diversify and enhance existing treatment options, particularly through combination regimens. The cAMP response element-binding protein (CREB) is a transcription factor involved in various biological processes. It is aberrantly activated in several aggressive cancer types, including breast cancer. Clinically, high CREB expression is associated with increased breast tumor aggressiveness and poor prognosis. Functionally, CREB promotes breast cancer cell proliferation, survival, invasion, metastasis, as well as therapy resistance by deregulating genes related to apoptosis, cell cycle, and metabolism. Targeting CREB with small molecule inhibitors has demonstrated promise in preclinical studies. This review summarizes the current understanding of CREB mechanisms and their potential as a therapeutic target. SIGNIFICANCE STATEMENT: cAMP response element-binding protein (CREB) is a master regulator of multiple biological processes, including neurodevelopment, metabolic regulation, and immune response. CREB is a putative proto-oncogene in breast cancer that regulates the cell cycle, apoptosis, and cellular migration. Preclinical development of CREB-targeting small molecules is underway.
Collapse
Affiliation(s)
- Jinghui Hong
- Department of Breast Surgery, General Surgery Centre, The First Hospital of Jilin University, Changchun, Jilin, China; Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Yuheng Wu
- Department of Breast Surgery, General Surgery Centre, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Mengxin Li
- Department of Breast Surgery, General Surgery Centre, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Ki-Fong Man
- Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Dong Song
- Department of Breast Surgery, General Surgery Centre, The First Hospital of Jilin University, Changchun, Jilin, China.
| | - Siang-Boon Koh
- Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom; University Hospitals Bristol and Weston, National Health Service (NHS) Foundation Trust, Bristol, United Kingdom.
| |
Collapse
|
8
|
Mazzitelli M, Kiritoshi T, Presto P, Hurtado Z, Antenucci N, Ji G, Neugebauer V. BDNF Signaling and Pain Modulation. Cells 2025; 14:476. [PMID: 40214430 PMCID: PMC11987912 DOI: 10.3390/cells14070476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is an important neuromodulator of nervous system functions and plays a key role in neuronal growth and survival, neurotransmission, and synaptic plasticity. The effects of BDNF are mainly mediated by the activation of tropomyosin receptor kinase B (TrkB), expressed in both the peripheral and central nervous system. BDNF has been implicated in several neuropsychiatric conditions such as schizophrenia and anxio-depressive disorders, as well as in pain states. This review summarizes the evidence for a critical role of BDNF throughout the pain system and describes contrasting findings of its pro- and anti-nociceptive effects. Different cellular sources of BDNF, its influence on neuroimmune signaling in pain conditions, and its effects in different cell types and regions are described. These and endogenous BDNF levels, downstream signaling mechanisms, route of administration, and approaches to manipulate BDNF functions could explain the bidirectional effects in pain plasticity and pain modulation. Finally, current knowledge gaps concerning BDNF signaling in pain are discussed, including sex- and pathway-specific differences.
Collapse
Affiliation(s)
- Mariacristina Mazzitelli
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (M.M.); (T.K.); (P.P.); (Z.H.); (N.A.); (G.J.)
| | - Takaki Kiritoshi
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (M.M.); (T.K.); (P.P.); (Z.H.); (N.A.); (G.J.)
| | - Peyton Presto
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (M.M.); (T.K.); (P.P.); (Z.H.); (N.A.); (G.J.)
| | - Zachary Hurtado
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (M.M.); (T.K.); (P.P.); (Z.H.); (N.A.); (G.J.)
| | - Nico Antenucci
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (M.M.); (T.K.); (P.P.); (Z.H.); (N.A.); (G.J.)
| | - Guangchen Ji
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (M.M.); (T.K.); (P.P.); (Z.H.); (N.A.); (G.J.)
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (M.M.); (T.K.); (P.P.); (Z.H.); (N.A.); (G.J.)
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
9
|
Esvald EE, Moistus A, Lehe K, Avarlaid A, Šubina A, Kuusemets L, Tuvikene J, Timmusk T. Stimulus-Dependent Expression of Bdnf Is Mediated by ATF2, MYT1L, and EGR1 Transcription Factors. J Neurosci 2025; 45:e0313242025. [PMID: 39947922 PMCID: PMC11924897 DOI: 10.1523/jneurosci.0313-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 01/23/2025] [Accepted: 01/31/2025] [Indexed: 03/21/2025] Open
Abstract
Neurotrophins like BDNF have a key role in the proper functioning of the central nervous system, influencing numerous processes like memory formation and behavior. An imbalance in BDNF levels can lead to a wide range of diseases, including depression and neurodevelopmental disorders. While the potential therapeutic effects of BDNF are well-recognized, there is a knowledge gap in understanding the mechanisms governing BDNF expression levels. Here, we focused on the regulation of Bdnf gene expression in response to different stimuli, specifically studying the effects of neuronal activity and BDNF-TrkB signaling on Bdnf transcription in cultured neurons from rats of either sex. We used in vitro DNA pulldown combined with mass spectrometry to determine transcription factors that interact with the Bdnf promoters upon different stimuli and validated numerous known regulators, such as USF and AP1 family, and novel candidate regulators using reporter assays. We show that the USF family of transcription factors is specifically recruited after membrane depolarization, whereas the AP1 family participates in Bdnf regulation only after BDNF-TrkB signaling. We further describe ATF2, MYT1L, and EGR family as novel regulators of Bdnf expression by demonstrating their direct binding to Bdnf promoters using chromatin immunoprecipitation assays both in vitro and in vivo, showing their functional role in Bdnf gene expression and ultimately identifying their regulatory cis-elements in Bdnf promoters. Furthermore, our results show competition between ATF2, CREB, and AP1 family in regulating Bdnf levels. Collectively, our results provide insight into the regulation of Bdnf expression upon different stimuli.
Collapse
Affiliation(s)
- Eli-Eelika Esvald
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn 12618, Estonia
- Protobios LLC, Tallinn 12618, Estonia
| | - Andra Moistus
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn 12618, Estonia
| | - Karin Lehe
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn 12618, Estonia
| | - Annela Avarlaid
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn 12618, Estonia
| | - Anastassia Šubina
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn 12618, Estonia
| | - Liis Kuusemets
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn 12618, Estonia
| | - Jürgen Tuvikene
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn 12618, Estonia
- Protobios LLC, Tallinn 12618, Estonia
- dxlabs LLC, Tallinn 12618, Estonia
| | - Tõnis Timmusk
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn 12618, Estonia
- Protobios LLC, Tallinn 12618, Estonia
| |
Collapse
|
10
|
Sun M, Qiu X, Yuan Z, Xu C, Chen Z. New advances in Traditional Chinese Medicine interventions for epilepsy: where are we and what do we know? Chin Med 2025; 20:37. [PMID: 40098198 PMCID: PMC11917061 DOI: 10.1186/s13020-025-01088-z] [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] [Accepted: 03/01/2025] [Indexed: 03/19/2025] Open
Abstract
Epilepsy, one of the most common neurological diseases, affects more than 70 million people worldwide. Anti-seizure drugs targeting membrane ion channels or GABAergic neurotransmission are the first choices for controlling seizures, whereas the high incidence of pharmacoresistance and adverse effects largely restrict the availability of current anti-seizure drugs (ASDs). Traditional Chinese Medicine (TCM) has shown historical evidence-based therapeutic effects for neurological diseases including epilepsy. But until the late 1990s, great efforts in both clinical and experimental fields advanced TCM interventions for epilepsy from evidence-based practices to more systematic neuropharmacological significance, and show new lights on preferable management of epilepsy in the last decade. This review summarized the advances of applying TCM interventions (ranging from herbal medicines and their active ingredients to other strategies such as acupuncture) for epilepsy, followed by associated mechanism theories. The therapeutic potential of TCM interventions for epilepsy as well as its comorbidities turns from somehow debatable to hopeful. Finally, some prospects and directions were proposed to drive further clinical translational research. The future directions of TCM should aim at not only deriving specific anti-epileptic molecules but also illustrating more precise mechanisms with the assistance of advanced multifaceted experimental tools.
Collapse
Affiliation(s)
- Minjuan Sun
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Huzhou Central Hospital, the Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiaoyun Qiu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Huzhou Central Hospital, the Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Zhijian Yuan
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Huzhou Central Hospital, the Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Cenglin Xu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Huzhou Central Hospital, the Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Huzhou Central Hospital, the Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| |
Collapse
|
11
|
Bocchieri E, Zimbone S, Giuffrida ML, Di Natale G, Sabatino G, Vecchio G, Pappalardo G, Chiechio S. Memantine and amantadine KLVFF peptide conjugates: Synthesis, structure determination, amyloid-β interaction and effects on recognition memory in mice. Eur J Pharmacol 2025; 990:177274. [PMID: 39848528 DOI: 10.1016/j.ejphar.2025.177274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Adamantane derivatives, such as memantine (Mem) and amantadine (Ada), have distinct mechanisms and therapeutic applications. Ada is primarily utilized as an antiviral and anti-Parkinson drug without significant pro-cognitive effects, Mem is effective in various clinical conditions characterized by cognitive deficits, including Alzheimer's disease. Recent evidence highlights a neuroprotective role for Aβ monomers, suggesting that preventing their aggregation into toxic oligomers could be a promising therapeutic strategy. Based on the observation that the Lys-Leu-Val-Phe-Phe (KLVFF) peptide, can block the transition of randomly coiled Aβ monomers into toxic β-sheet aggregates, two KLVFF conjugates, the Mem-Succ-KLVFF and Ada-Succ-KLVFF were investigated. METHODS Peptides were synthesized by Microwave-Assisted Solid Phase Peptide Synthesis (MW-SPPS). Circular Dichroism (CD), Th-T fluorescence and Gel-Electrophoresis techniques were used to assess the inhibitory effect on Aβ42 fibrillogenesis. The formation of inclusion complexes with β-Cyclodextrin (β-CyD) was demonstrated by NMR Spectroscopy. The Novel Object Recognition (NOR) test, followed by double-blind analysis, was applied for in vivo response to compounds administration. In vitro effects on neurons were studied by MTT assay and WB analysis, whereas HR ESI-MS allowed the molecular detection on brain homogenates. RESULTS These compounds differently affect Aβ42 aggregation. Mem-Succ-KLVFF, and Succ-KLVFF affect pCREB levels in differentiated SH-SY5Y, a signaling pathway involved in memory processes. In the NOR test, both Mem and KLVFF exhibited pro-cognitive effects individually and synergistically when co-administered. CONCLUSION Structure-activity relationships are discussed, integrating in vivo results, memory-related cellular pathways, and HR-ESI-MS analyses. These findings support the therapeutic potential of these compounds in preserving cognitive function.
Collapse
Affiliation(s)
- Eleonora Bocchieri
- Department of Drug and Health Sciences, Pharmacology and Toxicology Section, University of Catania, Italy; Institute of Crystallography, National Research Council (CNR-IC), 95126, Catania, Italy
| | - Stefania Zimbone
- Institute of Crystallography, National Research Council (CNR-IC), 95126, Catania, Italy
| | - Maria Laura Giuffrida
- Institute of Crystallography, National Research Council (CNR-IC), 95126, Catania, Italy
| | - Giuseppe Di Natale
- Institute of Crystallography, National Research Council (CNR-IC), 95126, Catania, Italy
| | - Giuseppina Sabatino
- Institute of Crystallography, National Research Council (CNR-IC), 95126, Catania, Italy
| | | | - Giuseppe Pappalardo
- Institute of Crystallography, National Research Council (CNR-IC), 95126, Catania, Italy.
| | - Santina Chiechio
- Department of Drug and Health Sciences, Pharmacology and Toxicology Section, University of Catania, Italy; Oasi Research Institute-IRCCS, 94018, Troina, Italy.
| |
Collapse
|
12
|
Yang X, Huang YWA, Marshall J. Targeting TrkB-PSD-95 coupling to mitigate neurological disorders. Neural Regen Res 2025; 20:715-724. [PMID: 38886937 PMCID: PMC11433911 DOI: 10.4103/nrr.nrr-d-23-02000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/15/2024] [Accepted: 03/30/2024] [Indexed: 06/20/2024] Open
Abstract
Tropomyosin receptor kinase B (TrkB) signaling plays a pivotal role in dendritic growth and dendritic spine formation to promote learning and memory. The activity-dependent release of brain-derived neurotrophic factor at synapses binds to pre- or postsynaptic TrkB resulting in the strengthening of synapses, reflected by long-term potentiation. Postsynaptically, the association of postsynaptic density protein-95 with TrkB enhances phospholipase Cγ-Ca2+/calmodulin-dependent protein kinase II and phosphatidylinositol 3-kinase-mechanistic target of rapamycin signaling required for long-term potentiation. In this review, we discuss TrkB-postsynaptic density protein-95 coupling as a promising strategy to magnify brain-derived neurotrophic factor signaling towards the development of novel therapeutics for specific neurological disorders. A reduction of TrkB signaling has been observed in neurodegenerative disorders, such as Alzheimer's disease and Huntington's disease, and enhancement of postsynaptic density protein-95 association with TrkB signaling could mitigate the observed deficiency of neuronal connectivity in schizophrenia and depression. Treatment with brain-derived neurotrophic factor is problematic, due to poor pharmacokinetics, low brain penetration, and side effects resulting from activation of the p75 neurotrophin receptor or the truncated TrkB.T1 isoform. Although TrkB agonists and antibodies that activate TrkB are being intensively investigated, they cannot distinguish the multiple human TrkB splicing isoforms or cell type-specific functions. Targeting TrkB-postsynaptic density protein-95 coupling provides an alternative approach to specifically boost TrkB signaling at localized synaptic sites versus global stimulation that risks many adverse side effects.
Collapse
Affiliation(s)
- Xin Yang
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Yu-Wen Alvin Huang
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
- Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, RI, USA
| | - John Marshall
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| |
Collapse
|
13
|
Shi C, Asaba S, Nakamura S, Matsui T. Ergothioneine Stimulates Ca 2+-Mediated Brain-Derived Neurotrophic Factor Expression in NE-4C Nerve Cells. ACS OMEGA 2025; 10:7004-7012. [PMID: 40028048 PMCID: PMC11866013 DOI: 10.1021/acsomega.4c09920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/02/2025] [Accepted: 02/05/2025] [Indexed: 03/05/2025]
Abstract
Ergothioneine (EGT), a naturally occurring histidine derivative, has been reported to modulate neurodegenerative diseases; however, the underlying mechanism remains unclear. This study aimed to investigate the brain-beneficial role of the natural amino acid EGT in NE-4C nerve cells. In the nerve cells, EGT treatment of >10 μM for 48 h significantly increased the expression of brain-derived neurotrophic factor (BDNF), as well as the phosphorylation of cAMP response element-binding protein (CREB), whereas no change was observed in acetylcholine receptor expression. Additionally, EGT induced an increase in intracellular Ca2+ levels via stimulation of the inositol 1,4,5-triphosphate receptor (IP3R) in the endoplasmic reticulum; this increase was abrogated by the inhibition of organic cation transporter 1 (OCTN1). Structure-activity relationship analysis revealed the importance of the trimethylammonium group in EGT for intracellular events. In conclusion, EGT incorporated into cells via the OCTN1 route may act as a nerve transmission stimulator via IP3R-mediated Ca2+-CREB/BDNF activation.
Collapse
Affiliation(s)
- Caiyue Shi
- Department of Bioscience
and Biotechnology, Faculty of Agriculture, Graduate School of Kyushu University, Fukuoka 819-0395, Japan
| | - Sumire Asaba
- Department of Bioscience
and Biotechnology, Faculty of Agriculture, Graduate School of Kyushu University, Fukuoka 819-0395, Japan
| | - Saya Nakamura
- Department of Bioscience
and Biotechnology, Faculty of Agriculture, Graduate School of Kyushu University, Fukuoka 819-0395, Japan
| | - Toshiro Matsui
- Department of Bioscience
and Biotechnology, Faculty of Agriculture, Graduate School of Kyushu University, Fukuoka 819-0395, Japan
| |
Collapse
|
14
|
Xu N, He Y, Wei YN, Bai L, Wang L. Possible antidepressant mechanism of acupuncture: targeting neuroplasticity. Front Neurosci 2025; 19:1512073. [PMID: 40018358 PMCID: PMC11865234 DOI: 10.3389/fnins.2025.1512073] [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: 10/16/2024] [Accepted: 02/03/2025] [Indexed: 03/01/2025] Open
Abstract
Major depressive disorder (MDD) is a highly prevalent and severely disabling psychiatric disorder that decreases quality of life and imposes substantial economic burden. Acupuncture has emerged as an effective adjunctive treatment for depression, it regulates neurotransmitters involved in mood regulation and modulates the activity of specific brain regions associated with emotional processing, as evidenced by neuroimaging and biochemical studies. Despite these insights, the precise neuroplastic mechanisms through which acupuncture exerts its antidepressant effects remain not fully elucidated. This review aims to summarize the current knowledge on acupuncture's modulation of neuroplasticity in depression, with a focus on the neuroplasticity-based targets associated with acupuncture's antidepressant effects. We encapsulate two decades of research into the neurobiological mechanisms underpinning the efficacy of acupuncture in treating depression. Additionally, we detail the acupoints and electroacupuncture parameters used in the treatment of depression to better serve clinical application.
Collapse
Affiliation(s)
- Ning Xu
- Department of First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yue He
- Department of First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yong-Nan Wei
- Department of First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lu Bai
- Department of First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Long Wang
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
15
|
Carrillo JÁ, Arcusa R, Xandri-Martínez R, Cerdá B, Zafrilla P, Marhuenda J. Impact of Polyphenol-Rich Nutraceuticals on Cognitive Function and Neuroprotective Biomarkers: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Nutrients 2025; 17:601. [PMID: 40004930 PMCID: PMC11858811 DOI: 10.3390/nu17040601] [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: 01/10/2025] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Recent studies have highlighted the neuroprotective effects of polyphenols, particularly their role in enhancing brain-derived neurotrophic factor (BDNF) and cAMP response element-binding protein (CREB) activity. This study aimed to evaluate the relationship between BDNF and CREB levels and cognitive performance in individuals undergoing a polyphenol-rich dietary intervention. METHODS A randomized, crossover, double-blind, placebo-controlled clinical trial was conducted with 92 participants. The intervention involved the daily intake of an encapsulated concentrate of fruit, vegetable, and berry juice powders (Juice Plus+ Premium®) over two 16-week periods, separated by a 4-week washout phase. Cognitive function was assessed using the Stroop Test, Trail Making Test, and Reynolds Intellectual Screening Test (RIST). The plasma levels of CREB and BDNF were measured using ELISA. RESULTS The polyphenol-rich product significantly improved cognitive performance, as evidenced by higher scores in the Stroop Test and RIST, compared to the placebo. Additionally, the plasma levels of CREB and BDNF were notably elevated in the product condition, indicating enhanced neuroprotective activity. CONCLUSIONS The findings suggest that polyphenol-rich nutraceuticals can modulate neurobiological mechanisms underlying cognitive improvements, primarily through the reduction of oxidative stress and the regulation of signaling pathways associated with synaptic plasticity. These results support the potential of dietary polyphenols in promoting cognitive health and preventing neurodegenerative diseases.
Collapse
Affiliation(s)
- Juan Ángel Carrillo
- Faculty of Pharmacy and Nutrition, Universidad Católica San Antonio, 30107 Murcia, Spain; (J.Á.C.); (R.A.); (B.C.)
| | - Raúl Arcusa
- Faculty of Pharmacy and Nutrition, Universidad Católica San Antonio, 30107 Murcia, Spain; (J.Á.C.); (R.A.); (B.C.)
| | | | - Begoña Cerdá
- Faculty of Pharmacy and Nutrition, Universidad Católica San Antonio, 30107 Murcia, Spain; (J.Á.C.); (R.A.); (B.C.)
| | - Pilar Zafrilla
- Faculty of Pharmacy and Nutrition, Universidad Católica San Antonio, 30107 Murcia, Spain; (J.Á.C.); (R.A.); (B.C.)
| | - Javier Marhuenda
- Faculty of Pharmacy and Nutrition, Universidad Católica San Antonio, 30107 Murcia, Spain; (J.Á.C.); (R.A.); (B.C.)
| |
Collapse
|
16
|
Aboul-Fotouh S, Zohny SM, Elnahas EM, Habib MZ, Hassan GA. Can memantine treat autism? Answers from preclinical and clinical studies. Neurosci Biobehav Rev 2025; 169:106019. [PMID: 39826825 DOI: 10.1016/j.neubiorev.2025.106019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/02/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Autism Spectrum Disorder (ASD) represents a clinical challenge due to its diverse behavioral symptoms and complex neuro-pathophysiology. Finding effective treatments that target the fundamental mechanisms of ASD remains a top priority. This narrative review presents the potential of the NMDA-receptor blocker memantine in managing ASD symptoms. Preclinical studies indicate that memantine could abrogate excitotoxicity, GABA/glutamate imbalance, reduced levels of brain-derived neurotrophic factor (BDNF), blood-brain barrier (BBB) leakage, and neuroinflammation, offering hope for managing core deficits associated with ASD like impaired social interaction and repetitive behaviors. However, clinical trials yield conflicting results, with some showing slight improvements in symptom severity and cognitive function, while others demonstrate limited efficacy. Further exploration of memantine's neurobiological mechanisms and refinement of treatment approaches are crucial for comprehensively tackling ASD complexities. Drawing from both animal models and clinical data, this review examines memantine's impact on core ASD symptoms, cognitive function, and potential mechanisms of action. Lastly, it identifies research gaps and proposes avenues for future investigations to enhance our understanding and utilization of memantine in ASD management.
Collapse
Affiliation(s)
- Sawsan Aboul-Fotouh
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sohir M Zohny
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Esraa M Elnahas
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mohamed Z Habib
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt; Basic Medical Sciences Department, Faculty of Medicine, King Salman International University, El Tor, South Sinai, Egypt.
| | - Ghada Am Hassan
- Neuropsychiatry Department, Faculty of Medicine, Galala University, Suez, Egypt; Neuropsychiatry Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
17
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [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
|
18
|
Garduño‐Tamayo NA, Almazán JL, Romo‐Rodríguez R, Valle‐García D, Meza‐Sosa KF, Pérez‐Domínguez M, Pelayo R, Pedraza‐Alva G, Pérez‐Martínez L. Klf10 Regulates the Emergence of Glial Phenotypes During Hypothalamic Development. J Neurosci Res 2025; 103:e70020. [PMID: 39924964 PMCID: PMC11808290 DOI: 10.1002/jnr.70020] [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: 11/28/2023] [Revised: 09/26/2024] [Accepted: 01/08/2025] [Indexed: 02/11/2025]
Abstract
Glial cells play a pivotal role in the Central Nervous System (CNS), constituting most brain cells. Gliogenesis, crucial in CNS development, occurs after neurogenesis. In the hypothalamus, glial progenitors first generate oligodendrocytes and later astrocytes. However, the precise molecular mechanisms governing the emergence of glial lineages in the developing hypothalamus remain incompletely understood. This study reveals the pivotal role of the transcription factor KLF10 in regulating the emergence of both astrocyte and oligodendrocyte lineages during embryonic hypothalamic development. Through transcriptomic and bioinformatic analyses, we identified novel KLF10 putative target genes, which play important roles in the differentiation of neurons, astrocytes, and oligodendrocytes. Notably, in the absence of KLF10, there is an increase in the oligodendrocyte population, while the astrocyte population decreases in the embryonic hypothalamus. Strikingly, this decline in the number of astrocytes persists into adulthood, indicating that the absence of KLF10 leads to an extended period of oligodendrocyte emergence while delaying the appearance of astrocytes. Our findings also unveil a novel signaling pathway for Klf10 gene expression regulation. We demonstrate that Klf10 is a target of CREB and that its expression is upregulated via the BDNF-p38-CREB pathway. Thus, we postulate that KLF10 is an integral part of the hypothalamic developmental program that ensures the correct timing for glial phenotypes' generation. Importantly, we propose that the Klf10-/- mouse model represents a valuable tool for investigating the impact of reduced astrocyte and microglia populations in the homeostasis of the adult hypothalamus.
Collapse
Affiliation(s)
- Norma Angelica Garduño‐Tamayo
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de BiotecnologíaUniversidad Nacional Autónoma de México (UNAM)CuernavacaMorelosMexico
| | - Jorge Luis Almazán
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de BiotecnologíaUniversidad Nacional Autónoma de México (UNAM)CuernavacaMorelosMexico
| | - Rubí Romo‐Rodríguez
- Laboratorio de Citómica del Cáncer Infantil, Centro de Investigación Biomédica de OrienteDelegación PueblaPueblaMexico
| | - David Valle‐García
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de BiotecnologíaUniversidad Nacional Autónoma de México (UNAM)CuernavacaMorelosMexico
| | - Karla F. Meza‐Sosa
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de BiotecnologíaUniversidad Nacional Autónoma de México (UNAM)CuernavacaMorelosMexico
| | - Martha Pérez‐Domínguez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de BiotecnologíaUniversidad Nacional Autónoma de México (UNAM)CuernavacaMorelosMexico
| | - Rosana Pelayo
- Laboratorio de Citómica del Cáncer Infantil, Centro de Investigación Biomédica de OrienteDelegación PueblaPueblaMexico
| | - Gustavo Pedraza‐Alva
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de BiotecnologíaUniversidad Nacional Autónoma de México (UNAM)CuernavacaMorelosMexico
| | - Leonor Pérez‐Martínez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de BiotecnologíaUniversidad Nacional Autónoma de México (UNAM)CuernavacaMorelosMexico
| |
Collapse
|
19
|
Zhang Y, Liu C, Zhu Q, Wu H, Liu Z, Zeng L. Relationship Between Depression and Epigallocatechin Gallate from the Perspective of Gut Microbiota: A Systematic Review. Nutrients 2025; 17:259. [PMID: 39861389 PMCID: PMC11767295 DOI: 10.3390/nu17020259] [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: 12/06/2024] [Revised: 01/07/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Depression, a serious mental illness, is characterized by high risk, high incidence, persistence, and tendency to relapse, posing a significant burden on global health. The connection between depression and gut microbiota is an emerging field of study in psychiatry and neuroscience. Understanding the gut-brain axis is pivotal for understanding the pathogenesis and treatment of depression. Gut microbes influence depression-like behaviors by impacting the hypothalamic-pituitary-adrenal axis (HPA), monoamine neurotransmitters, immune responses, cell signaling, and metabolic pathways. Tea, widely used in clinical practice to improve neuropsychiatric disorders, contains Epigallocatechin gallate (EGCG), a major ingredient of green tea, which effectively regulates intestinal flora. This review examined the risks and causes of depression, the complications associated with intestinal flora, their role in the development and treatment of depression, and how EGCG may alleviate depression through interactions with gut microbiota and other mechanisms.
Collapse
Affiliation(s)
- Yangbo Zhang
- School of Pharmacy, Shaoyang University, Shaoyang 422000, China; (Y.Z.); (Q.Z.); (H.W.)
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China;
| | - Changwei Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China;
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Qi Zhu
- School of Pharmacy, Shaoyang University, Shaoyang 422000, China; (Y.Z.); (Q.Z.); (H.W.)
| | - Hui Wu
- School of Pharmacy, Shaoyang University, Shaoyang 422000, China; (Y.Z.); (Q.Z.); (H.W.)
| | - Zhonghua Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China;
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China
- Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China
| | - Li Zeng
- School of Pharmacy, Shaoyang University, Shaoyang 422000, China; (Y.Z.); (Q.Z.); (H.W.)
| |
Collapse
|
20
|
Abdelaziz HA, Hamed MF, Ghoniem HA, Nader MA, Suddek GM. Empagliflozin Mitigates PTZ-Induced Seizures in Rats: Modulating Npas4 and CREB-BDNF Signaling Pathway. J Neuroimmune Pharmacol 2025; 20:5. [PMID: 39776284 PMCID: PMC11706855 DOI: 10.1007/s11481-024-10162-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025]
Abstract
Empagliflozin (EMPA) is one of the sodium/glucose cotransporter 2 (SGLT2) inhibitors that has been recently approved for the treatment of diabetes mellitus type II. Recently, EMPA has shown protective effects in different neurological disorders, besides its antidiabetic activity. Kindling is a relevant model to study epilepsy and neuroplasticity. This study aimed to investigate the potential protective effects of EMPA (1 and 3 mg/kg orally) against convulsant effects induced by pentylenetetrazole (PTZ) using a modified window- (win-) PTZ kindling protocol. The biochemical dysfunction and hippocampal damage induced by PTZ were profoundly reversed by EMPA treatment in a dose-dependent manner, as evidenced by the significant increase in reduced glutathione (GSH) and decrease in malondialdehyde (MDA) hippocampal contents. Furthermore, EMPA counteracted PTZ-induced neuronal damage in the hippocampal region, as confirmed by histopathological examination of the hippocampal tissues. EMPA impaired astrocytosis and showed an antiapoptotic effect through a significant reduction of glial fibrillary acidic protein (GFAP) and BCL2-Associated X Protein (BAX) expressions, respectively. Interestingly, EMPA exhibited an antiepileptic effect against PTZ-induced seizures through significantly reducing neuronal PAS domain Protein 4 (Npas4), cyclic adenosine monophosphate (cAMP) response element binding protein (CREB) hippocampal expressions, and enhancing the brain-derived neurotrophic factor (BDNF)-tropomyosin receptor kinase B (TrkB) pathway, which are found to be involved in epileptogenesis, eventually leading to significant improvement of behavioral impairments induced by PTZ. Hence, these results showed further prospective insights for EMPA as a neuroprotective agent.
Collapse
Affiliation(s)
- Heba A Abdelaziz
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 35712, Egypt
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Mohamed F Hamed
- Pathology Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Hamdy A Ghoniem
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Manar A Nader
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura National University, Gamasa, 7731168, Egypt
| | - Ghada M Suddek
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| |
Collapse
|
21
|
Deabes DAH, El-Abd EAW, Baraka SM, El-Gendy ZA, Korany RMS, Elbatanony MM. Metabolomics analyses and comparative insight to neuroprotective potential of unripe fruits and leaves of Citrus aurantium ethanolic extracts against cadmium-induced rat brain dysfunction: involvement of oxidative stress and akt-mediated CREB/BDNF and GSK3β/NF-κB signaling pathways. Metab Brain Dis 2025; 40:89. [PMID: 39760898 PMCID: PMC11703990 DOI: 10.1007/s11011-024-01513-6] [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: 08/09/2024] [Accepted: 12/17/2024] [Indexed: 01/07/2025]
Abstract
Serious neurological disorders were associated with cadmium toxicity. Hence, this research aimed to investigate the potential neuroprotective impacts of the ethanolic extracts of Citrus aurantium unripe fruits and leaves (CAF and CAL, respectively) at doses 100 and 200 mg/kg against cadmium chloride-provoked brain dysfunction in rats for 30 consecutive days. HPLC for natural pigment content revealed that CAF implied higher contents of Chlorophyll B, while the CAL has a high yield of chlorophyll A and total carotenoid. Fifty-seven chromatographic peaks were identified by UPLC/MS/MS; 49 and 29 were recognized from CAF or CAL, respectively. Four compounds were isolated from CAF: 3',4',7 -trihydroxyflavone, isorhainetin, vitexin, and apigenin. In vitro studies outlined the antioxidant capacity of studied extracts where CAF showed better scavenging radical DPPH activity. Results clarified that both extracts with a superior function of CAF at the high adopted dose significantly ameliorated CdCl2-induced neuro-oxidative stress and neuro-inflammatory response via restoring antioxidant status and hindering nuclear factor kappa B (NF-κB) stimulation. Moreover, it up-regulated the levels of phospho-protein kinase B (p-Akt), phospho- cAMP-response element binding protein (p-CREB), and brain-derived neurotropic factor (BDNF) levels, and elicited a marked decrease in the content of glycogen synthase kinase 3 beta (GSK3β), besides amending Caspase-3 and hyperphosphorylation of tau protein in brain tissues. Moreover, a significant improvement in the rats' behavioral tasks of the CAL and CAF-treated groups has been recorded, as indicated by marked preservation in locomotion, exploratory, and memory functions of the experimental rats. In conclusion, the reported neuroprotective impacts of C. aurantium extracts may be through modulating p-AKT/p-CREB/BDNF and / or p-Akt/ GSK3β/NF-κB signaling pathways.
Collapse
Affiliation(s)
- Doaa A H Deabes
- Pharmacognosy Department, National Research Centre (NRC), El Behouth St., P.O. 12622, Cairo, Egypt
| | - Eman A W El-Abd
- Pharmacognosy Department, National Research Centre (NRC), El Behouth St., P.O. 12622, Cairo, Egypt
| | - Sara M Baraka
- Chemistry of Natural Compounds Department, National Research Centre, Giza, 12622, Egypt.
| | - Zeinab A El-Gendy
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt
| | - Reda M S Korany
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Marwa M Elbatanony
- Pharmacognosy Department, National Research Centre (NRC), El Behouth St., P.O. 12622, Cairo, Egypt
| |
Collapse
|
22
|
Chen Y, Mao L, Zhou Q, Bai D, Kong Y. Role of BDNF-TrkB signaling in the improvement of motor function and neuroplasticity after ischemic stroke in rats by transcranial direct current stimulation. Brain Res Bull 2025; 220:111164. [PMID: 39662631 DOI: 10.1016/j.brainresbull.2024.111164] [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: 06/13/2024] [Revised: 11/14/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Transcranial direct current stimulation (tDCS) has an impact on improving cognitive and motor dysfunction induced by ischemia-reperfusion injury. However, to use this technology more rationally in clinical practice, a deepened understanding of the molecular mechanisms behind its therapeutic effects is needed. This study explored the role of the brain-derived neurotrophic factor(BDNF) and its associated receptor tropomyosin-receptor kinase B(TrkB) while deciphering the underlying mechanisms in transcranial direct current therapy to treat ischemic stroke. METHODS A middle cerebral artery occlusion-reperfusion(MCAO/R) model was established in rats to observe tDCS effects on brain damage. Behavioral tests, the modified neurologic severity score(mNSS), and the Hoffman reflex / the M wave(Hmax/Mmax) ratio helped assess motor function and neurologic deficits. HE and Nissl staining helped observe the morphological changes and count of nerve cells. We tested the expression of growth-associated protein-43(Gap-43) and microtubule-associated protein-2(Map-2), K+-Cl- co-transporter 2(KCC2), γ-aminobutyric acid(GABA), and key BDNF-TrkB downstream signaling, the phospholipase C gamma(PLCγ) / CaMK IV / cAMP response element binding protein(CREB), and extracellular signal-regulated protein kinase(ERK1/2) / ribosomal S6 kinase(RSK) using western blotting. Moreover, BDNF was analyzed in plasma using the enzyme-linked immunosorbent assay (ELISA) to investigate the tDCS effect on human BDNF expression levels. Finally, a BDNF receptor antagonist, ANA-12, was administered to explore the tDCS mechanism mediating BDNF-TrkB signaling. RESULTS After tDCS treatment, the mNSS was improved, and the motor function was restored. Moreover, tDCS decreased cell swelling after MCAO/R and enhanced the number of neurons. tDCS treatment increased: (1) BDNF, Gap-43, Map-2 expression, (2) KCC2, GABA, and (3) PLCγ, CaMK IV, CREB and ERK1/2, RSK. Furthermore, ELISA results indicate that tDCS elevated human plasma BDNF protein expression. However, the therapeutic effect of tDCS was suppressed to a certain extent by adding ANA-12. CONCLUSION Our findings indicate that tDCS may exert a neuroprotective effect by activating the downstream key molecules of BDNF-TrkB expression, for instance, PLCγ/ CaMK IV/ CREB and ERK/ RSK pathway. Moreover, tDCS can control neuronal excitability, promote axonal regeneration, and accelerate motor function recovery in ischemia reperfusion-injured rats.
Collapse
Affiliation(s)
- Yu Chen
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Respiratory and Critical Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan Province 610000, China
| | - Lin Mao
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Qinxiang Zhou
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Dingqun Bai
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Yuhan Kong
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
23
|
Li JZ, Ramalingam N, Li S. Targeting epigenetic mechanisms in amyloid-β-mediated Alzheimer's pathophysiology: unveiling therapeutic potential. Neural Regen Res 2025; 20:54-66. [PMID: 38767476 PMCID: PMC11246147 DOI: 10.4103/nrr.nrr-d-23-01827] [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: 11/07/2023] [Revised: 01/03/2024] [Accepted: 02/07/2024] [Indexed: 05/22/2024] Open
Abstract
Alzheimer's disease is a prominent chronic neurodegenerative condition characterized by a gradual decline in memory leading to dementia. Growing evidence suggests that Alzheimer's disease is associated with accumulating various amyloid-β oligomers in the brain, influenced by complex genetic and environmental factors. The memory and cognitive deficits observed during the prodromal and mild cognitive impairment phases of Alzheimer's disease are believed to primarily result from synaptic dysfunction. Throughout life, environmental factors can lead to enduring changes in gene expression and the emergence of brain disorders. These changes, known as epigenetic modifications, also play a crucial role in regulating the formation of synapses and their adaptability in response to neuronal activity. In this context, we highlight recent advances in understanding the roles played by key components of the epigenetic machinery, specifically DNA methylation, histone modification, and microRNAs, in the development of Alzheimer's disease, synaptic function, and activity-dependent synaptic plasticity. Moreover, we explore various strategies, including enriched environments, exposure to non-invasive brain stimulation, and the use of pharmacological agents, aimed at improving synaptic function and enhancing long-term potentiation, a process integral to epigenetic mechanisms. Lastly, we deliberate on the development of effective epigenetic agents and safe therapeutic approaches for managing Alzheimer's disease. We suggest that addressing Alzheimer's disease may require distinct tailored epigenetic drugs targeting different disease stages or pathways rather than relying on a single drug.
Collapse
Affiliation(s)
- Jennie Z. Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Nagendran Ramalingam
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Shaomin Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
24
|
Zheng Y, Yu X, Wei L, Chen Q, Xu Y, Ni P, Deng W, Guo W, Hu X, Qi X, Li T. LT-102, an AMPA receptor potentiator, alleviates depression-like behavior and synaptic plasticity impairments in prefrontal cortex induced by sleep deprivation. J Affect Disord 2024; 367:18-30. [PMID: 39214374 DOI: 10.1016/j.jad.2024.08.176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Sleep loss is closely related to the onset and development of depression, and the mechanisms involved may include impaired synaptic plasticity. Considering the important role of glutamate α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate receptors (AMPARs) in synaptic plasticity as well as depression, we introduce LT-102, a novel AMPARs potentiator, to evaluate the potential of LT-102 in treating sleep deprivation-induced depression-like behaviors. METHODS We conducted a comprehensive behavioral assessment to evaluate the effects of LT-102 on depression-like symptoms in male C57BL/6J mice. This assessment included the open field test to measure general locomotor activity and anxiety-like behavior, the forced swimming test and tail suspension test to assess despair behaviors indicative of depressive states, and the sucrose preference test to quantify anhedonia, a core symptom of depression. Furthermore, to explore the impact of LT-102 on synaptic plasticity, we utilized a combination of Western blot analysis to detect protein expression levels, Golgi-Cox staining to visualize neuronal morphology, and immunofluorescence to examine the localization of synaptic proteins. Additionally, we utilized primary cortical neurons to delineate the signaling pathway modulated by LT-102. RESULTS Treatment with LT-102 significantly reduced depression-like behaviors associated with sleep deprivation. Quantitative Western blot (WB) analysis revealed a significant increase in GluA1 phosphorylation in the prefrontal cortex (PFC), triggering the Ca2+/calmodulin-dependent protein kinase II/cAMP response element-binding protein/brain-derived neurotrophic factor (CaMKII/CREB/BDNF) and forkhead box protein P2/postsynaptic density protein 95 (FoxP2/PSD95) signaling pathways. Immunofluorescence imaging confirmed that LT-102 treatment increased spine density and co-labeling of PSD95 and vesicular glutamate transporter 1 (VGLUT1) in the PFC, reversing the reductions typically observed following sleep deprivation. Golgi staining further validated these results, showing a substantial increase in neuronal dendritic spine density in sleep-deprived mice treated with LT-102. Mechanistically, application of LT-102 to primary cortical neurons, resulted in elevated levels of phosphorylated AKT (p-AKT) and phosphorylated glycogen synthase kinase-3 beta (p-GSK3β), key downstream molecules in the BDNF signaling pathway, which in turn upregulated FoxP2 and PSD95 expression. LIMITATIONS In our study, we chose to exclusively use male mice to eliminate potential influences of the estrous cycle on behavior and physiology. As there is no widely accepted positive drug control for sleep deprivation studies, we did not include one in our research. CONCLUSION Our results suggest that LT-102 is a promising therapeutic agent for counteracting depression-like behaviors and synaptic plasticity deficits induced by sleep deprivation, primarily through the activation of CaMKII/CREB/BDNF and AKT/GSK3β/FoxP2/PSD95 signaling pathways.
Collapse
Affiliation(s)
- Yanghao Zheng
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-computer Interface Institute, Hangzhou 311100, China
| | - Xueli Yu
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-computer Interface Institute, Hangzhou 311100, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Long Wei
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qiyuan Chen
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yan Xu
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Peiyan Ni
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-computer Interface Institute, Hangzhou 311100, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China
| | - Wei Deng
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-computer Interface Institute, Hangzhou 311100, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China
| | - Wanjun Guo
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-computer Interface Institute, Hangzhou 311100, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China
| | - Xun Hu
- The Clinical Research Center and Department of Pathology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xueyu Qi
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-computer Interface Institute, Hangzhou 311100, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China.
| | - Tao Li
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-computer Interface Institute, Hangzhou 311100, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
25
|
Jakkamsetti MS, Kolusu AS, Rongala S, Arakareddy BP, Nori LP, Samudrala PK. Saroglitazar, a PPAR α/γ agonist alleviates 3-Nitropropionic acid induced neurotoxicity in rats: Unveiling the underlying mechanisms. Neurotoxicology 2024; 105:131-146. [PMID: 39326639 DOI: 10.1016/j.neuro.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
Saroglitazar (SGZ), a peroxisomal proliferated activated receptor α/γ agonist showed neuroprotective effects in various neurodegenerative disorders like Alzheimer's and Parkinson's. However, no studies were performed on Huntington's, so the goal of the current study is to examine the effect of SGZ on Huntington's disease like symptoms induced by 3-Nitropropionic acid. In this protocol, twenty-four rats were divided into four groups, each group consisting of 6 animals. Group 1: The control group received 1 % CMC 10 mg/kg, p.o. for 14 days. Groups 2, 3, and 4 received 3-NP 15 mg/kg, i.p. from Day 1 to Day 7. Groups 3 and 4 received SGZ 5 mg/kg, p.o. and 10 mg/kg, p.o. respectively once daily from day 1 to day 14. Various behavioral tests like OFT, rotarod, hanging wire, narrow beam walk, MWM, and Y-maze were performed. On day-15, the animals were euthanised by cervical dislocation and brain sample were isolated for biochemical and histopathological analysis. Administration of 3-NP showed a significant decrease in motor coordination and cognitive function. Furthermore, 3-NP altered the activity of acetylcholinesterase, anti-oxidant enzymes, Nrf-2, NF-κB, BDNF, CREB levels, and histological features. However, treatment with SGZ showed ameliorative effects in the 3-NP induced neurotoxicity via PPAR α/γ pathway by reducing motor dysfunction, memory impairment, cholinesterase levels, oxidative stress, neuroinflammation. It also enhanced the levels of Nrf-2, BDNF, and CREB expression and improved histological features. In conclusion, treatment with Saroglitazar attenuated Huntington's disease-like symptoms in rats which are induced by 3-NP via activation of PPAR α/γ pathway.
Collapse
Affiliation(s)
- Madhuri Suma Jakkamsetti
- Department of Pharmacology, Shri Vishnu College of Pharmacy (SVCP), Vishnupur, Bhimavaram, West Godavari, Andhra Pradesh 534202, India
| | - Aravinda Sai Kolusu
- Department of Pharmacology, Shri Vishnu College of Pharmacy (SVCP), Vishnupur, Bhimavaram, West Godavari, Andhra Pradesh 534202, India
| | - Suma Rongala
- Department of Pharmacology, Shri Vishnu College of Pharmacy (SVCP), Vishnupur, Bhimavaram, West Godavari, Andhra Pradesh 534202, India
| | - Bhanu Prakash Arakareddy
- Department of Pharmacology, Shri Vishnu College of Pharmacy (SVCP), Vishnupur, Bhimavaram, West Godavari, Andhra Pradesh 534202, India
| | - Lakshmi Prashanthi Nori
- Department of Pharmaceutics, Shri Vishnu College of Pharmacy (SVCP), Vishnupur, Bhimavaram, West Godavari, Andhra Pradesh 534202, India
| | - Pavan Kumar Samudrala
- Department of Pharmacology, Shri Vishnu College of Pharmacy (SVCP), Vishnupur, Bhimavaram, West Godavari, Andhra Pradesh 534202, India.
| |
Collapse
|
26
|
Muhammad RN, Albahairy MA, Abd El Fattah MA, Ibrahim WW. Empagliflozin-activated AMPK elicits neuroprotective properties in reserpine-induced depression via regulating dynamics of hippocampal autophagy/inflammation and PKCζ-mediated neurogenesis. Psychopharmacology (Berl) 2024; 241:2565-2584. [PMID: 39158617 PMCID: PMC11569022 DOI: 10.1007/s00213-024-06663-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/29/2024] [Indexed: 08/20/2024]
Abstract
RATIONALE Major depression has been an area of extensive research during the last decades, for it represents a leading cause of disability and suicide. The stark rise of depression rates influenced by life stressors, economic threats, pandemic era, and resistance to classical treatments, has made the disorder rather challenging. Adult hippocampal neurogenesis and plasticity are particularly sensitive to the dynamic interplay between autophagy and inflammation. In fact, the intricate balance between the two processes contributes to neuronal homeostasis and survival. OBJECTIVES Having demonstrated promising potentials in AMPK activation, a major metabolic sensor and autophagy regulator, empagliflozin (Empa) was investigated for possible antidepressant properties in the reserpine rat model of depression. RESULTS While the reserpine protocol elicited behavioral, biochemical, and histopathological changes relevant to depression, Empa outstandingly hindered these pathological perturbations. Importantly, hippocampal autophagic response markedly declined with reserpine which disrupted the AMPK/mTOR/Beclin1/LC3B machinery and, conversely, neuro-inflammation prevailed under the influence of the NLRP3 inflammasome together with oxidative/nitrative stress. Consequently, AMPK-mediated neurotrophins secretion obviously deteriorated through PKCζ/NF-κB/BDNF/CREB signal restriction. Empa restored hippocampal monoamines and autophagy/inflammation balance, driven by AMPK activation. By promoting the atypical PKCζ phosphorylation (Thr403) which subsequently phosphorylates NF-κB at Ser311, AMPK successfully reinforced BDNF/CREB signal and hippocampal neuroplasticity. The latter finding was supported by hippocampal CA3 toluidine blue staining to reveal intact neurons. CONCLUSION The current study highlights an interesting role for Empa as a regulator of autophagic and inflammatory responses in the pathology of depression. The study also pinpoints an unusual contribution for NF-κB in neurotrophins secretion via AMPK/PKCζ/NF-κB/BDNF/CREB signal transduction. Accordingly, Empa can have special benefits in diabetic patients with depressive symptoms. LIMITATIONS The influence of p-NF-κB (Ser311) on NLRP3 inflammasome assembly and activation has not been investigated, which can represent an interesting point for further research.
Collapse
Affiliation(s)
- Radwa N Muhammad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Mohammed A Albahairy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Mai A Abd El Fattah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Weam W Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
27
|
Kim M, Lee H, Kwon S, Cho S, Um MY. Phlorotannin Supplement Improves Scopolamine-Induced Memory Dysfunction by Rescuing Synaptic Damage in Mice. J Microbiol Biotechnol 2024; 34:2301-2309. [PMID: 39317682 PMCID: PMC11637834 DOI: 10.4014/jmb.2407.07009] [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: 07/05/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/26/2024]
Abstract
This study investigated the efficacy of a phlorotannin supplement (PS) in ameliorating scopolamine (SCO)-induced memory deficits in mice, focusing on synaptic function and the underlying molecular mechanisms. Male C57BL/6N mice were divided into six groups and treated with vehicle, donepezil (5 mg/kg body weight, (BW)), or PS (100, 250, or 500 mg/kg BW) for 6 weeks. Behavioral tests were conducted, followed by Golgi staining, immunofluorescence, and immunoblotting to assess synaptic protein expression and signaling pathways. Behavioral tests showed that PS administration significantly improved SCO-induced memory impairment and restored synaptic protein expression (synaptophysin, synapsin1, and postsynaptic density protein 95) in the hippocampus. Additionally, PS enhanced brain-derived neurotrophic factor (BDNF) signaling and activated the extracellular signal-regulated kinase/CAMP response element binding protein (ERK-CREB) pathway, essential for synaptic plasticity. Our findings demonstrate that PS mitigates SCO-induced memory dysfunction by protecting synaptic integrity and activating the BDNF-ERK-CREB signaling pathway, indicating the potential of PS as a natural intervention for treating memory deficits.
Collapse
Affiliation(s)
- Minji Kim
- Division of Functional Food Research, Korea Food Research Institute, Wanju 55365, Republic of Korea
- Department of Food Biotechnology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Haeun Lee
- Division of Functional Food Research, Korea Food Research Institute, Wanju 55365, Republic of Korea
- Department of Food Biotechnology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Sangoh Kwon
- S&D Research and Development Institute, Cheongju 28156 Republic of Korea
| | - Seungmok Cho
- Department of Food Science and Technology/Institute of Food Science, Pukyong National University, Busan 48513, Republic of Korea
| | - Min Young Um
- Division of Functional Food Research, Korea Food Research Institute, Wanju 55365, Republic of Korea
- Department of Food Biotechnology, University of Science and Technology, Daejeon 34113, Republic of Korea
| |
Collapse
|
28
|
Sung KC, Wang LY, Wang CC, Chu CH, Sun HS, Hsiao YH. Enhanced hippocampal TIAM2S expression alleviates cognitive deficits in Alzheimer's disease model mice. Pharmacol Rep 2024; 76:1032-1043. [PMID: 39012419 DOI: 10.1007/s43440-024-00623-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/17/2024]
Abstract
BACKGROUND Dendritic spine dysfunction is a key feature of Alzheimer's disease (AD) pathogenesis. Human T-cell lymphoma invasion and metastasis 2 (TIAM2) is expressed in two isoforms, the full length (TIAM2L) and a short transcript (TIAM2S). Compared to TIAM2L protein, which is undetectable, TIAM2S protein is abundant in human brain tissue, especially the hippocampus, and can promote neurite outgrowth in our previous findings. However, whether enhanced hippocampal TIAM2S expression can alleviate cognitive deficits in Alzheimer's disease model mice remains unclear. METHODS We crossbred 3xTg-AD with TIAM2S mice to generate an AD mouse model that carries the human TIAM2S gene (3xTg-AD/TIAM2S mice). The Morris water maze and object location tests assessed hippocampus-dependent spatial memory. Lentiviral-driven shRNA or cDNA approaches were used to manipulate hippocampal TIAM2S expression. Golgi staining and Sholl analysis were utilized to measure neuronal dendrites and dendritic spines in the mouse hippocampi. RESULTS Compared to 3xTg-AD mice, 3xTg-AD/TIAM2S mice displayed improved cognitive functions. According to the hippocampus is one of the earliest affected brain regions by AD, we further injected TIAM2S shRNA or TIAM2S cDNA into mouse hippocampi to confirm whether manipulating hippocampal TIAM2S expression could affect AD-related cognitive functions. The results showed that the reduced hippocampal TIAM2S expression in 3xTg-AD/TIAM2S mice abolished the memory improvement effect, whereas increased hippocampal TIAM2S levels alleviated cognitive deficits in 3xTg-AD mice. Furthermore, we found that TIAM2S-mediated memory improvement was achieved by regulating dendritic plasticity. CONCLUSIONS These results will provide new insights into connecting TIAM2S with AD and support the notion that TIAM2S should be investigated as potential AD therapeutic targets.
Collapse
Affiliation(s)
- Kuan-Chin Sung
- Department of Neurosurgery, Chi-Mei Medical Center, 901 Chung Hwa Road, Yung Kang City, Tainan, Taiwan
| | - Li-Yun Wang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Che-Chuan Wang
- Department of Neurosurgery, Chi-Mei Medical Center, 901 Chung Hwa Road, Yung Kang City, Tainan, Taiwan
| | - Chun-Hsien Chu
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - H Sunny Sun
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Hsin Hsiao
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
29
|
Tsybko A, Eremin D, Ilchibaeva T, Khotskin N, Naumenko V. CDNF Exerts Anxiolytic, Antidepressant-like, and Procognitive Effects and Modulates Serotonin Turnover and Neuroplasticity-Related Genes. Int J Mol Sci 2024; 25:10343. [PMID: 39408672 PMCID: PMC11482483 DOI: 10.3390/ijms251910343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/17/2024] [Accepted: 09/21/2024] [Indexed: 10/19/2024] Open
Abstract
Cerebral dopamine neurotrophic factor (CDNF) is an unconventional neurotrophic factor because it does not bind to a known specific receptor on the plasma membrane and functions primarily as an unfolded protein response (UPR) regulator in the endoplasmic reticulum. Data on the effects of CDNF on nonmotor behavior and monoamine metabolism are limited. Here, we performed the intracerebroventricular injection of a recombinant CDNF protein at doses of 3, 10, and 30 μg in C57BL/6 mice. No adverse effects of the CDNF injection on feed and water consumption or locomotor activity were observed for 3 days afterwards. Decreases in body weight and sleep duration were transient. CDNF-treated animals demonstrated improved performance on the operant learning task and a substantial decrease in anxiety and behavioral despair. CDNF in all the doses enhanced serotonin (5-HT) turnover in the murine frontal cortex, hippocampus, and midbrain. This alteration was accompanied by changes in the mRNA levels of the 5-HT1A and 5-HT7 receptors and in monoamine oxidase A mRNA and protein levels. We found that CDNF dramatically increased c-Fos mRNA levels in all investigated brain areas but elevated the phosphorylated-c-Fos level only in the midbrain. Similarly, enhanced CREB phosphorylation was found in the midbrain in experimental animals. Additionally, the upregulation of a spliced transcript of XBP1 (UPR regulator) was detected in the midbrain and frontal cortex. Thus, we can hypothesize that exogenous CDNF modulates the UPR pathway and overall neuronal activation and enhances 5-HT turnover, thereby affecting learning and emotion-related behavior.
Collapse
Affiliation(s)
- Anton Tsybko
- The Federal Research Center, Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia; (D.E.); (T.I.); (N.K.); (V.N.)
| | | | | | | | | |
Collapse
|
30
|
Wang Y, Peng D, Zhang X, Chen J, Feng J, Zhang R, Mai W, Chen H, Yang Y, Huang Y, Zhang Q. PLCβ4 driven by cadmium-exposure during gestation and lactation contributes to cognitive deficits by suppressing PIP2/PLCγ1/CREB/BDNF signaling pathway in male offspring. JOURNAL OF HAZARDOUS MATERIALS 2024; 474:134756. [PMID: 38820747 DOI: 10.1016/j.jhazmat.2024.134756] [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: 01/29/2024] [Revised: 05/13/2024] [Accepted: 05/27/2024] [Indexed: 06/02/2024]
Abstract
The fetus and infants are particularly vulnerable to Cadmium (Cd) due to the immaturity of the blood-brain barrier. In utero and early life exposure to Cd is associated with cognitive deficits. Although such exposure has attracted widespread attention, its gender-specificity remains controversial, and there are no reports disclosing the underlying mechanism of gender‑specific neurotoxicity. We extensively evaluated the learning and cognitive functions and synaptic plasticity of male and female rats exposed to maternal Cd. Maternal Cd exposure induced learning and memory deficits in male offspring rats, but not in female offspring rats. PLCβ4 was identified as a critical protein, which might be related to the gender‑specific cognitive deficits in male rats. The up-regulated PLCβ4 competed with PLCγ1 to bind to PIP2, which counteracted the hydrolysis of PIP2 by PLCγ1. The decreased activation of PLCγ1 inhibited the phosphorylation of CREB to reduce BDNF transcription, which consequently resulted in the damage of hippocampal neurons and cognitive deficiency. Moreover, the low level of BDNF promoted AEP activation to induce Aβ deposition in the hippocampus. These findings highlight that PLCβ4 might be a potential target for the therapy of learning and cognitive deficits caused by Cd exposure in early life.
Collapse
Affiliation(s)
- Youjin Wang
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Dong Peng
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Xiang Zhang
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Jiayan Chen
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Jianfeng Feng
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Runze Zhang
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Wanwen Mai
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Hongxia Chen
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China; National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou 510632, China
| | - Yan Yang
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China; National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou 510632, China
| | - Yadong Huang
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China; National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou 510632, China.
| | - Qihao Zhang
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China; National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
31
|
Chen J, Zhang Q, Xu W, Li Z, Chen X, Luo Q, Wang D, Peng L. Baicalein upregulates macrophage TREM2 expression via TrKB-CREB1 pathway to attenuate acute inflammatory injury in acute-on-chronic liver failure. Int Immunopharmacol 2024; 139:112685. [PMID: 39047449 DOI: 10.1016/j.intimp.2024.112685] [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: 02/07/2024] [Revised: 06/06/2024] [Accepted: 07/11/2024] [Indexed: 07/27/2024]
Abstract
OBJECTIVE Acute-on-chronic liver failure (ACLF) is a syndrome characterized by a high short-term mortality rate, and effective interventions are still lacking. This study aims to investigate whether the small molecule baicalein can mitigate ACLF and elucidate the molecular mechanisms. METHODS The ACLF mouse model was induced through chronic liver injury using carbon tetrachloride, followed by acute inflammation induction with lipopolysaccharide (LPS). Baicalein was administered through intraperitoneal injection to explore its therapeutic effects. In vitro experiments utilized the iBMDM macrophage cell line to investigate the underlying mechanisms. Peripheral blood was collected from clinical ACLF patients for validation. RESULTS In the LPS-induced ACLF mouse model, baicalein demonstrated a significant reduction in acute inflammation and liver damage, as evidenced by histopathological evaluation, liver function analysis, and inflammatory marker measurements. Transcriptomic analysis, coupled with molecular biology experiments, uncovered that baicalein exerts its effects in ACLF by activating the TrKB-CREB1 signaling axis to upregulate the surface expression of the TREM2 receptor on macrophages. This promotes M2 macrophage polarization and activates efferocytosis, thereby inhibiting inflammation and alleviating liver damage. Furthermore, we observed a substantial negative correlation between postoperative peripheral blood plasma soluble TREM2 (sTREM2) levels and inflammation, as well as adverse outcomes in clinical ACLF patients. CONCLUSION Baicalein plays a protective role in ACLF by enhancing the surface expression of the TREM2 receptor on macrophages, leading to the suppression of inflammation, mitigation of liver damage, and a reduction in mortality. Additionally, plasma sTREM2 emerges as a critical indicator for predicting adverse outcomes in ACLF patients.
Collapse
Affiliation(s)
- Jia Chen
- Department of Infectious Diseases and Guangdong Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qiongchi Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China; Department of Orthopedics, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Wenxiong Xu
- Department of Infectious Diseases and Guangdong Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhipeng Li
- Department of Infectious Diseases and Guangdong Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiyao Chen
- Department of Infectious Diseases and Guangdong Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qiumin Luo
- Department of Infectious Diseases and Guangdong Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dong Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China; Department of Orthopedics, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.
| | - Liang Peng
- Department of Infectious Diseases and Guangdong Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
32
|
Gross J, Knipper M, Mazurek B. Candidate Key Proteins in Tinnitus-A Bioinformatic Study of Synaptic Transmission in the Cochlear Nucleus. Biomedicines 2024; 12:1615. [PMID: 39062188 PMCID: PMC11274367 DOI: 10.3390/biomedicines12071615] [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: 06/04/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
The aim of this study was to identify key proteins of synaptic transmission in the cochlear nucleus (CN) that are involved in normal hearing, acoustic stimulation, and tinnitus. A gene list was compiled from the GeneCards database using the keywords "synaptic transmission" AND "tinnitus" AND "cochlear nucleus" (Tin). For comparison, two gene lists with the keywords "auditory perception" (AP) AND "acoustic stimulation" (AcouStim) were built. The STRING protein-protein interaction (PPI) network and the Cytoscape data analyzer were used to identify the top two high-degree proteins (HDPs) and their high-score interaction proteins (HSIPs), together referred to as key proteins. The top1 key proteins of the Tin-process were BDNF, NTRK1, NTRK3, and NTF3; the top2 key proteins are FOS, JUN, CREB1, EGR1, MAPK1, and MAPK3. Highly significant GO terms in CN in tinnitus were "RNA polymerase II transcription factor complex", "late endosome", cellular response to cadmium ion", "cellular response to reactive oxygen species", and "nerve growth factor signaling pathway", indicating changes in vesicle and cell homeostasis. In contrast to the spiral ganglion, where important changes in tinnitus are characterized by processes at the level of cells, important biological changes in the CN take place at the level of synapses and transcription.
Collapse
Affiliation(s)
- Johann Gross
- Tinnitus Center, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany;
- Leibniz Society of Science Berlin, 10117 Berlin, Germany;
| | - Marlies Knipper
- Leibniz Society of Science Berlin, 10117 Berlin, Germany;
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, 72076 Tübingen, Germany
| | - Birgit Mazurek
- Tinnitus Center, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany;
| |
Collapse
|
33
|
Woo MS, Bal LC, Winschel I, Manca E, Walkenhorst M, Sevgili B, Sonner JK, Di Liberto G, Mayer C, Binkle-Ladisch L, Rothammer N, Unger L, Raich L, Hadjilaou A, Noli B, Manai AL, Vieira V, Meurs N, Wagner I, Pless O, Cocco C, Stephens SB, Glatzel M, Merkler D, Friese MA. The NR4A2/VGF pathway fuels inflammation-induced neurodegeneration via promoting neuronal glycolysis. J Clin Invest 2024; 134:e177692. [PMID: 39145444 PMCID: PMC11324305 DOI: 10.1172/jci177692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 06/11/2024] [Indexed: 08/16/2024] Open
Abstract
A disturbed balance between excitation and inhibition (E/I balance) is increasingly recognized as a key driver of neurodegeneration in multiple sclerosis (MS), a chronic inflammatory disease of the central nervous system. To understand how chronic hyperexcitability contributes to neuronal loss in MS, we transcriptionally profiled neurons from mice lacking inhibitory metabotropic glutamate signaling with shifted E/I balance and increased vulnerability to inflammation-induced neurodegeneration. This revealed a prominent induction of the nuclear receptor NR4A2 in neurons. Mechanistically, NR4A2 increased susceptibility to excitotoxicity by stimulating continuous VGF secretion leading to glycolysis-dependent neuronal cell death. Extending these findings to people with MS (pwMS), we observed increased VGF levels in serum and brain biopsies. Notably, neuron-specific deletion of Vgf in a mouse model of MS ameliorated neurodegeneration. These findings underscore the detrimental effect of a persistent metabolic shift driven by excitatory activity as a fundamental mechanism in inflammation-induced neurodegeneration.
Collapse
Affiliation(s)
- Marcel S. Woo
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lukas C. Bal
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ingo Winschel
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elias Manca
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Biomedical Sciences, NEF-Laboratory, University of Cagliari, Monserrato, Cagliari, Italy
| | - Mark Walkenhorst
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bachar Sevgili
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jana K. Sonner
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Giovanni Di Liberto
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Christina Mayer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lars Binkle-Ladisch
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Rothammer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lisa Unger
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lukas Raich
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexandros Hadjilaou
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Protozoa Immunology, Bernhard-Nocht-Institute for Tropical Medicine (BNITM), Hamburg, Germany
| | - Barbara Noli
- Department of Biomedical Sciences, NEF-Laboratory, University of Cagliari, Monserrato, Cagliari, Italy
| | - Antonio L. Manai
- Department of Biomedical Sciences, NEF-Laboratory, University of Cagliari, Monserrato, Cagliari, Italy
| | - Vanessa Vieira
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nina Meurs
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ingrid Wagner
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Ole Pless
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Hamburg, Germany
| | - Cristina Cocco
- Department of Biomedical Sciences, NEF-Laboratory, University of Cagliari, Monserrato, Cagliari, Italy
| | - Samuel B. Stephens
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa, USA
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Doron Merkler
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Manuel A. Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
34
|
Sun Y, Xiao Z, Chen B, Zhao Y, Dai J. Advances in Material-Assisted Electromagnetic Neural Stimulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2400346. [PMID: 38594598 DOI: 10.1002/adma.202400346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/26/2024] [Indexed: 04/11/2024]
Abstract
Bioelectricity plays a crucial role in organisms, being closely connected to neural activity and physiological processes. Disruptions in the nervous system can lead to chaotic ionic currents at the injured site, causing disturbances in the local cellular microenvironment, impairing biological pathways, and resulting in a loss of neural functions. Electromagnetic stimulation has the ability to generate internal currents, which can be utilized to counter tissue damage and aid in the restoration of movement in paralyzed limbs. By incorporating implanted materials, electromagnetic stimulation can be targeted more accurately, thereby significantly improving the effectiveness and safety of such interventions. Currently, there have been significant advancements in the development of numerous promising electromagnetic stimulation strategies with diverse materials. This review provides a comprehensive summary of the fundamental theories, neural stimulation modulating materials, material application strategies, and pre-clinical therapeutic effects associated with electromagnetic stimulation for neural repair. It offers a thorough analysis of current techniques that employ materials to enhance electromagnetic stimulation, as well as potential therapeutic strategies for future applications.
Collapse
Affiliation(s)
- Yuting Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| |
Collapse
|
35
|
Abdelkawy YS, Elharoun M, Sheta E, Abdel-Raheem IT, Nematalla HA. Liraglutide and Naringenin relieve depressive symptoms in mice by enhancing Neurogenesis and reducing inflammation. Eur J Pharmacol 2024; 971:176525. [PMID: 38561101 DOI: 10.1016/j.ejphar.2024.176525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 03/11/2024] [Accepted: 03/21/2024] [Indexed: 04/04/2024]
Abstract
Depression is a debilitating mental disease that negatively impacts individuals' lives and society. Novel hypotheses have been recently proposed to improve our understanding of depression pathogenesis. Impaired neuroplasticity and upregulated neuro-inflammation add-on to the disturbance in monoamine neurotransmitters and therefore require novel anti-depressants to target them simultaneously. Recent reports demonstrate the antidepressant effect of the anti-diabetic drug liraglutide. Similarly, the natural flavonoid naringenin has shown both anti-diabetic and anti-depressant effects. However, the neuro-pharmacological mechanisms underlying their actions remain understudied. The study aims to evaluate the antidepressant effects and neuroprotective mechanisms of liraglutide, naringenin or a combination of both. Depression was induced in mice by administering dexamethasone (32 mcg/kg) for seven consecutive days. Liraglutide (200 mcg/kg), naringenin (50 mg/kg) and a combination of both were administered either simultaneously or after induction of depression for twenty-eight days. Behavioral and molecular assays were used to assess the progression of depressive symptoms and biomarkers. Liraglutide and naringenin alone or in combination alleviated the depressive behavior in mice, manifested by decrease in anxiety, anhedonia, and despair. Mechanistically, liraglutide and naringenin improved neurogenesis, decreased neuroinflammation and comparably restored the monoamines levels to that of the reference drug escitalopram. The drugs protected mice from developing depression when given simultaneously with dexamethasone. Collectively, the results highlight the usability of liraglutide and naringenin in the treatment of depression in mice and emphasize the different pathways that contribute to the pathogenesis of depression.
Collapse
Affiliation(s)
- Yara S Abdelkawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour 22514, Egypt
| | - Mona Elharoun
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour 22514, Egypt
| | - Eman Sheta
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria 21131, Egypt
| | - Ihab Talat Abdel-Raheem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour 22514, Egypt
| | - Hisham A Nematalla
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour 22514, Egypt.
| |
Collapse
|
36
|
Guha S, Nguyen AM, Young A, Mondell E, Farber DB. Decreased CREB phosphorylation impairs embryonic retinal neurogenesis in the Oa1-/- mouse model of Ocular albinism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.594013. [PMID: 38798688 PMCID: PMC11118284 DOI: 10.1101/2024.05.14.594013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Mutations in the human Ocular albinism type-1 gene OA1 are associated with abnormal retinal pigment epithelium (RPE) melanogenesis and poor binocular vision resulting from misrouting of ipsilateral retinal ganglion cell (iRGC) axons to the brain. We studied the latter using wild-type (WT) and Oa1-/- mouse eyes. At embryonic stages, the WT RPE-specific Oa1 protein signals through cAMP/Epac1-Erk2-CREB. Following CREB phosphorylation, a pCREB gradient extends from the RPE to the differentiating retinal amacrine and RGCs. In contrast to WT, the Oa1-/- RPE and ventral ciliary-margin-zone, a niche for iRGCs, express less pCREB while their retinas have a disrupted pCREB gradient, indicating Oa1's involvement in pCREB maintenance. Oa1-/- retinas also show hyperproliferation, enlarged nuclei, reduced differentiation, and fewer newborn amacrine and RGCs than WT retinas. Our results demonstrate that Oa1's absence leads to reduced binocular vision through a hyperproliferation-associated block in differentiation that impairs neurogenesis. This may affect iRGC axon's routing to the brain.
Collapse
Affiliation(s)
- Sonia Guha
- Department of Ophthalmology, Stein Eye Institute, UCLA School of Medicine, Los Angeles, CA 90095, USA
| | - Andrew M. Nguyen
- Department of Ophthalmology, Stein Eye Institute, UCLA School of Medicine, Los Angeles, CA 90095, USA
| | - Alejandra Young
- Department of Ophthalmology, Stein Eye Institute, UCLA School of Medicine, Los Angeles, CA 90095, USA
| | - Ethan Mondell
- Department of Ophthalmology, Stein Eye Institute, UCLA School of Medicine, Los Angeles, CA 90095, USA
| | - Debora B. Farber
- Department of Ophthalmology, Stein Eye Institute, UCLA School of Medicine, Los Angeles, CA 90095, USA
| |
Collapse
|
37
|
Cai XM, Sun XY, Li R, Wang PJ, Qiu JC, Ge YX, Yang L. The hippocampal salt-inducible kinase 2-CREB-regulated transcription co-activator 1 system mediates the antidepressant actions of paroxetine in mice. Behav Brain Res 2024; 465:114972. [PMID: 38552744 DOI: 10.1016/j.bbr.2024.114972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/13/2024] [Accepted: 03/24/2024] [Indexed: 04/06/2024]
Abstract
The hippocampal salt-inducible kinase 2 (SIK2)-CREB-regulated transcription co-activator 1 (CRTC1) system has been demonstrated to participate in not only the pathogenesis of depression but also the antidepressant mechanisms of several antidepressant medications including fluoxetine, paroxetine, and mirtazapine. Like fluoxetine, paroxetine is also a widely used selective serotonin (5-HT) reuptake inhibitor (SSRI). Recent studies have indicated that paroxetine also modulates several pharmacological targets other than the 5-HT system. Here, we speculate that paroxetine regulates the hippocampal SIK2-CRTC1 system. Chronic stress models of depression, various behavioral tests, western blotting, co-immunoprecipitation, quantitative real-time reverse transcription PCR, and genetic knockdown were used together in the present study. Our results show that the antidepressant actions of paroxetine in mice models of depression were accompanied by its preventing effects against chronic stress on hippocampal SIK2, CRTC1, and CRTC1-CREB binding. In contrast, genetic knockdown of hippocampal CRTC1 notably abrogated the antidepressant effects of paroxetine in mice. In summary, regulating hippocampal SIK2 and CRTC1 participates in the antidepressant mechanism of paroxetine, extending the knowledge of its pharmacological targets.
Collapse
Affiliation(s)
- Xiang-Ming Cai
- Department of Pharmacy, Nantong Fourth People's Hospital, Nantong, Jiangsu 226000, China
| | - Xiao-Yu Sun
- Department of Pharmacy, Nantong Fourth People's Hospital, Nantong, Jiangsu 226000, China
| | - Rui Li
- Department of Pharmacy, Nantong Fourth People's Hospital, Nantong, Jiangsu 226000, China
| | - Pei-Juan Wang
- Department of Psychiatry, Nantong Fourth People's Hospital, Nantong, Jiangsu 226000, China
| | - Jian-Cheng Qiu
- Department of Psychiatry, Nantong Fourth People's Hospital, Nantong, Jiangsu 226000, China
| | - Yu-Xin Ge
- Department of Pharmacy, Nantong Fourth People's Hospital, Nantong, Jiangsu 226000, China
| | - Lei Yang
- Department of Pharmacy, Nantong Fourth People's Hospital, Nantong, Jiangsu 226000, China.
| |
Collapse
|
38
|
Yang Y, Hang W, Li J, Liu T, Hu Y, Fang F, Yan D, McQuillan PM, Wang M, Hu Z. Effect of General Anesthetic Agents on Microglia. Aging Dis 2024; 15:1308-1328. [PMID: 37962460 PMCID: PMC11081156 DOI: 10.14336/ad.2023.1108] [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: 09/07/2023] [Accepted: 11/08/2023] [Indexed: 11/15/2023] Open
Abstract
The effects of general anesthetic agents (GAAs) on microglia and their potential neurotoxicity have attracted the attention of neuroscientists. Microglia play important roles in the inflammatory process and in neuromodulation of the central nervous system. Microglia-mediated neuroinflammation is a key mechanism of neurocognitive dysfunction during the perioperative period. Microglial activation by GAAs induces anti-inflammatory and pro-inflammatory effects in microglia, suggesting that GAAs play a dual role in the mechanism of postoperative cognitive dysfunction. Understanding of the mechanisms by which GAAs regulate microglia may help to reduce the incidence of postoperative adverse effects. Here, we review the actions of GAAs on microglia and the consequent changes in microglial function. We summarize clinical and animal studies associating microglia with general anesthesia and describe how GAAs interact with neurons via microglia to further explore the mechanisms of action of GAAs in the nervous system.
Collapse
Affiliation(s)
- Yanchang Yang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Wenxin Hang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jun Li
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Anesthesiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, China.
| | - Tiantian Liu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Anesthesiology, Ningbo Women and Children's Hospital, Ningbo, China.
| | - Yuhan Hu
- Cell Biology Department, Yale University, New Haven, CT, USA.
| | - Fuquan Fang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Dandan Yan
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Patrick M. McQuillan
- Department of Anesthesiology, Penn State Hershey Medical Center, Penn State College of Medicine, Hershey, PA, USA.
| | - Mi Wang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Zhiyong Hu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
39
|
Ning B, Ge T, Wu Y, Wang Y, Zhao M. Role of Brain-Derived Neurotrophic Factor in Anxiety or Depression After Percutaneous Coronary Intervention. Mol Neurobiol 2024; 61:2921-2937. [PMID: 37946008 DOI: 10.1007/s12035-023-03758-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023]
Abstract
Anxiety or depression after percutaneous coronary intervention (PCI) is one of the key clinical problems in cardiology that need to be solved urgently. Brain-derived neurotrophic factor (BDNF) may be a potential biomarker for the pathogenesis and treatment of anxiety or depression after PCI. This article reviews the correlation between BDNF and cardiovascular system and nervous system from the aspects of synthesis, release and action site of BDNF, and focuses on the latest research progress of the mechanism of BDNF in anxiety or depression after PCI. It includes the specific mechanisms by which BDNF regulates the levels of inflammatory factors, reduces oxidative stress damage, and mediates multiple signaling pathways. In addition, this review summarizes the therapeutic potential of BDNF as a potential biomarker for anxiety or depression after PCI.
Collapse
Affiliation(s)
- Bo Ning
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Teng Ge
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Yongqing Wu
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Yuting Wang
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
- Affiliated Hospital, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Mingjun Zhao
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China.
- Affiliated Hospital, Shaanxi University of Chinese Medicine, Xianyang, 712046, China.
- Shaanxi Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Cardiovascular Diseases, Xianyang, 712046, China.
| |
Collapse
|
40
|
Song S, Oft H, Metwally S, Paruchuri S, Bielanin J, Fiesler V, Sneiderman C, Kohanbash G, Sun D. Deletion of Slc9a1 in Cx3cr1 + cells stimulated microglial subcluster CREB1 signaling and microglia-oligodendrocyte crosstalk. J Neuroinflammation 2024; 21:69. [PMID: 38509618 PMCID: PMC10953158 DOI: 10.1186/s12974-024-03065-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 03/15/2024] [Indexed: 03/22/2024] Open
Abstract
Microglial Na/H exchanger-1 (NHE1) protein, encoded by Slc9a1, plays a role in white matter demyelination of ischemic stroke brains. To explore underlying mechanisms, we conducted single cell RNA-seq transcriptome analysis in conditional Slc9a1 knockout (cKO) and wild-type (WT) mouse white matter tissues at 3 days post-stroke. Compared to WT, Nhe1 cKO brains expanded a microglial subgroup with elevated transcription of white matter myelination genes including Spp1, Lgals3, Gpnmb, and Fabp5. This subgroup also exhibited more acidic pHi and significantly upregulated CREB signaling detected by ingenuity pathway analysis and flow cytometry. Moreover, the Nhe1 cKO white matter tissues showed enrichment of a corresponding oligodendrocyte subgroup, with pro-phagocytosis and lactate shuffling gene expression, where activated CREB signaling is a likely upstream regulator. These findings demonstrate that attenuation of NHE1-mediated H+ extrusion acidifies microglia/macrophage and may underlie the stimulation of CREB1 signaling, giving rise to restorative microglia-oligodendrocyte interactions for remyelination.
Collapse
Affiliation(s)
- Shanshan Song
- Department of Neurology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA.
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, USA.
- Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15213, USA.
| | - Helena Oft
- Department of Neurology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shamseldin Metwally
- Department of Neurology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, USA
| | - Satya Paruchuri
- Department of Neurology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, USA
| | - John Bielanin
- Department of Neurology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, USA
| | - Victoria Fiesler
- Department of Neurology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chaim Sneiderman
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gary Kohanbash
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA.
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, USA.
- Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
41
|
Park K, Kong CH, Kang WC, Jeon M, Lee WH, Lee J, Kim SC, Jung SY, Ryu JH. LPC20K modified from krill oil ameliorates the scopolamine-induced cognitive impairment. Behav Brain Res 2024; 461:114836. [PMID: 38145873 DOI: 10.1016/j.bbr.2023.114836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 12/22/2023] [Accepted: 12/22/2023] [Indexed: 12/27/2023]
Abstract
Alzheimer's disease (AD) is characterized by cognitive impairment. It is common in the elderly. Etiologically, dysfunction of cholinergic neurotransmitter system is prominent in AD. However, disease modifying drug for AD is still unavailable. We hypothesized that krill oil and modified krill oil containing 20 % lysophosphatidylcholine-docosahexaenoic acid (LPC-DHA, LPC20K) could play a crucial role in AD by improving cognitive functions measured by several behavioral tests. We found that LPC20K could ameliorate short-term, long-term, spatial, and object recognition memory under cholinergic hypofunction states. To find the underlying mechanism involved in the effect of LPC20K on cognitive function, we investigated changes of signaling molecules using Western blotting. Expression levels of protein kinase C zeta (PKCζ) and postsynaptic density protein 95 (PSD-95), and phosphorylation levels of extracellular signal-regulated kinase (ERK), Ca2+/calmodulin-dependent protein kinase Ⅱ (CaMKⅡ), and cAMP response element-binding protein (CREB) were significantly increased in LPC20K-administered group compared to those in the memory impairment group. Moreover, the expression levels of BDNF were temporally increased especially 6 or 9 h after administration of LPC20K compared with the control group. These results suggest that LPC20K could ameliorate memory impairment caused by hypocholinergic state by enhancing the expression levels of PKCζ and PSD-95, and phosphorylation levels of ERK, CaMKⅡ and CREB and increasing BDNF expression levels. Therefore, LPC20K could be used as a dietary supplement against cognitive impairment observed in diseases such as AD with a hypocholinergic state.
Collapse
Affiliation(s)
- Keontae Park
- Department of Biomedical and Pharmaceutical Science, Kyung Hee University, Seoul 02447, the Republic of Korea
| | - Chang Hyeon Kong
- Department of Biomedical and Pharmaceutical Science, Kyung Hee University, Seoul 02447, the Republic of Korea
| | - Woo Chang Kang
- Department of Biomedical and Pharmaceutical Science, Kyung Hee University, Seoul 02447, the Republic of Korea
| | - Mijin Jeon
- Department of Biomedical and Pharmaceutical Science, Kyung Hee University, Seoul 02447, the Republic of Korea
| | - Won Hyung Lee
- Department of Biomedical and Pharmaceutical Science, Kyung Hee University, Seoul 02447, the Republic of Korea
| | - Juyeon Lee
- Croda Korea Ltd., Seongnam-si, Gyeonggi-do 13636, the Republic of Korea
| | - Sang Chul Kim
- Croda Korea Ltd., Seongnam-si, Gyeonggi-do 13636, the Republic of Korea
| | - Seo Yun Jung
- Department of Biomedical and Pharmaceutical Science, Kyung Hee University, Seoul 02447, the Republic of Korea
| | - Jong Hoon Ryu
- Department of Biomedical and Pharmaceutical Science, Kyung Hee University, Seoul 02447, the Republic of Korea; Department of Oriental Pharmaceutical Science, Kyung Hee University, Seoul 02447, the Republic of Korea.
| |
Collapse
|
42
|
Jiang ZF, Xuan LN, Sun XW, Liu SB, Yin J. Knockdown of SIK3 in the CA1 Region can Reduce Seizure Susceptibility in Mice by Inhibiting Decreases in GABA AR α1 Expression. Mol Neurobiol 2024; 61:1404-1416. [PMID: 37715891 DOI: 10.1007/s12035-023-03630-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 08/30/2023] [Indexed: 09/18/2023]
Abstract
Imbalance between excitation and inhibition is an important cause of epilepsy. Salt-inducible kinase 1 (SIK1) gene mutation can cause epilepsy. In this study, we first found that the expression of SIK3 is increased after epilepsy. Furthermore, the role of SIK3 in epilepsy was explored. In cultured hippocampal neurons, we used Pterosin B, a selective SIK3 inhibitor that can inhibit epileptiform discharges induced by the convulsant drug cyclothiazide (a positive allosteric modulator of AMPA receptors, CTZ). Knockdown of SIK3 inhibited epileptiform discharges and increased the amplitude of miniature inhibitory postsynaptic currents (mIPSCs). In mice, knockdown of SIK3 reduced epilepsy susceptibility in a pentylenetetrazole (a GABAA receptor antagonist, PTZ) acute kindling experiment and increased the expression of GABAA receptor α1. In conclusion, our results suggest that blockade or knockdown of SIK3 can inhibit epileptiform discharges and that SIK3 has the potential to be a novel target for epilepsy treatment.
Collapse
Affiliation(s)
- Zhen-Fu Jiang
- Dalian Medical University, Dalian, 116044, Liaoning, China.
- Department of Neurosurgery, the Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Shahekou, Dalian, 116023, Liaoning, China.
| | - Li-Na Xuan
- Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Xiao-Wan Sun
- East China Normal University, Shanghai, 200241, China
| | - Shao-Bo Liu
- Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Jian Yin
- Dalian Medical University, Dalian, 116044, Liaoning, China.
- Department of Neurosurgery, the Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Shahekou, Dalian, 116023, Liaoning, China.
| |
Collapse
|
43
|
Zhang Y, Li S, Li L, Huang H, Fu Z, Hua Z. Bilirubin impairs neuritogenesis and synaptogenesis in NSPCs by downregulating NMDAR-CREB-BDNF signaling. In Vitro Cell Dev Biol Anim 2024; 60:161-171. [PMID: 38216855 DOI: 10.1007/s11626-023-00844-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 12/01/2023] [Indexed: 01/14/2024]
Abstract
Neonatal jaundice is one of the most common disorders in the first 2 wk after birth. Unconjugated bilirubin (UCB) is neurotoxic and can cause neurological dysfunction; however, the underlying mechanisms remain unclear. Neurogenesis, neuronal growth, and synaptogenesis are exuberant in the early postnatal stage. In this study, the impact of UCB on neuritogenesis and synaptogenesis in the early postnatal stage was evaluated both in vitro and in vivo. Primary culture neuronal stem and progenitor cells (NSPCs) were treated with UCB during differentiation, and then the neurite length and synapse puncta were measured. In the bilirubin encephalopathy (BE) animal model, DCX+-marked developing neurons were used to detect apical length and dendritic arborization. According to the data, UCB significantly reduced neurite length and synapse density, as well as decreased the apical dendrite length and dendritic arborization. Furthermore, the NMDAR subunit NR2B was downregulated in NSPCs, while pCREB expression in the hippocampus progressively decreased during disease progression in the BE model. Next, we tested the expression of NR2B, pCREB, mBDNF, and p-mTOR in NSPCs in vitro, and found that UCB treatment reduced the expression of these proteins. In summary, this suggests that UCB causes chronic neurological impairment and is related to the inhibition of NMDAR-CREB-BDNF signaling in NSPCs, which is associated with reduced neuritogenesis and synaptogenesis. This finding may inspire the development of novel pharmaceuticals and treatments.
Collapse
Affiliation(s)
- Yan Zhang
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Siyu Li
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Ling Li
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Hongmei Huang
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Zhou Fu
- Department of Respiratory Diseases, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| | - Ziyu Hua
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| |
Collapse
|
44
|
McEwan AR, Hing B, Erickson JC, Hutchings G, Urama C, Norton-Hughes E, D'Ippolito M, Berry S, Delibegovic M, Grassmann F, MacKenzie A. An ancient polymorphic regulatory region within the BDNF gene associated with obesity modulates anxiety-like behaviour in mice and humans. Mol Psychiatry 2024; 29:660-670. [PMID: 38228888 PMCID: PMC11153140 DOI: 10.1038/s41380-023-02359-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/10/2023] [Accepted: 12/01/2023] [Indexed: 01/18/2024]
Abstract
Obesity and anxiety are morbidities notable for their increased impact on society during the recent COVID-19 pandemic. Understanding the mechanisms governing susceptibility to these conditions will increase our quality of life and resilience to future pandemics. In the current study, we explored the function of a highly conserved regulatory region (BE5.1) within the BDNF gene that harbours a polymorphism strongly associated with obesity (rs10767664; p = 4.69 × 10-26). Analysis in primary cells suggested that the major T-allele of BE5.1 was an enhancer, whereas the obesity-associated A-allele was not. However, CRISPR/CAS9 deletion of BE5.1 from the mouse genome (BE5.1KO) produced no significant effect on the expression of BDNF transcripts in the hypothalamus, no change in weight gain after 28 days and only a marginally significant increase in food intake. Nevertheless, transcripts were significantly increased in the amygdala of female mice and elevated zero maze and marble-burying tests demonstrated a significant increase in anxiety-like behaviour that could be reversed by diazepam. Consistent with these observations, human GWAS cohort analysis demonstrated a significant association between rs10767664 and anxiousness in human populations. Intriguingly, interrogation of the human GTEx eQTL database demonstrated no effect on BDNF mRNA levels associated with rs10767664 but a highly significant effect on BDNF-antisense (BDNF-AS) gene expression and splicing. The subsequent observation that deletion of BE5.1 also significantly reduced BDNF-AS expression in mice suggests a novel mechanism in the regulation of BDNF expression common to mice and humans, which contributes to the modulation of mood and anxiety in both species.
Collapse
Affiliation(s)
- Andrew R McEwan
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB24 2ZD, UK
| | - Benjamin Hing
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Johanna C Erickson
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB24 2ZD, UK
| | - Greg Hutchings
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB24 2ZD, UK
| | - Charity Urama
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB24 2ZD, UK
| | - Emily Norton-Hughes
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB24 2ZD, UK
| | - Mariam D'Ippolito
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB24 2ZD, UK
| | - Susan Berry
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB24 2ZD, UK
| | - Mirela Delibegovic
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB24 2ZD, UK
| | - Felix Grassmann
- Institute for Clinical Research and Systems Medicine, Health and Medical University, Potsdam, Germany
| | - Alasdair MacKenzie
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB24 2ZD, UK.
| |
Collapse
|
45
|
Liu S, Zhong M, Wu H, Su W, Wang Y, Li P. Potential Beneficial Effects of Naringin and Naringenin on Long COVID-A Review of the Literature. Microorganisms 2024; 12:332. [PMID: 38399736 PMCID: PMC10892048 DOI: 10.3390/microorganisms12020332] [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: 01/09/2024] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused a severe epidemic due to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Recent studies have found that patients do not completely recover from acute infections, but instead, suffer from a variety of post-acute sequelae of SARS-CoV-2 infection, known as long COVID. The effects of long COVID can be far-reaching, with a duration of up to six months and a range of symptoms such as cognitive dysfunction, immune dysregulation, microbiota dysbiosis, myalgic encephalomyelitis/chronic fatigue syndrome, myocarditis, pulmonary fibrosis, cough, diabetes, pain, reproductive dysfunction, and thrombus formation. However, recent studies have shown that naringenin and naringin have palliative effects on various COVID-19 sequelae. Flavonoids such as naringin and naringenin, commonly found in fruits and vegetables, have various positive effects, including reducing inflammation, preventing viral infections, and providing antioxidants. This article discusses the molecular mechanisms and clinical effects of naringin and naringenin on treating the above diseases. It proposes them as potential drugs for the treatment of long COVID, and it can be inferred that naringin and naringenin exhibit potential as extended long COVID medications, in the future likely serving as nutraceuticals or clinical supplements for the comprehensive alleviation of the various manifestations of COVID-19 complications.
Collapse
Affiliation(s)
- Siqi Liu
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-Evaluation of Post-Market Traditional Chinese Medicine, State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (S.L.); (M.Z.); (H.W.); (W.S.); (Y.W.)
| | - Mengli Zhong
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-Evaluation of Post-Market Traditional Chinese Medicine, State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (S.L.); (M.Z.); (H.W.); (W.S.); (Y.W.)
| | - Hao Wu
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-Evaluation of Post-Market Traditional Chinese Medicine, State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (S.L.); (M.Z.); (H.W.); (W.S.); (Y.W.)
| | - Weiwei Su
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-Evaluation of Post-Market Traditional Chinese Medicine, State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (S.L.); (M.Z.); (H.W.); (W.S.); (Y.W.)
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China
| | - Yonggang Wang
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-Evaluation of Post-Market Traditional Chinese Medicine, State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (S.L.); (M.Z.); (H.W.); (W.S.); (Y.W.)
| | - Peibo Li
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-Evaluation of Post-Market Traditional Chinese Medicine, State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (S.L.); (M.Z.); (H.W.); (W.S.); (Y.W.)
| |
Collapse
|
46
|
Dinevska M, Widodo SS, Cook L, Stylli SS, Ramsay RG, Mantamadiotis T. CREB: A multifaceted transcriptional regulator of neural and immune function in CNS tumors. Brain Behav Immun 2024; 116:140-149. [PMID: 38070619 DOI: 10.1016/j.bbi.2023.12.002] [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: 07/06/2023] [Revised: 11/16/2023] [Accepted: 12/04/2023] [Indexed: 01/21/2024] Open
Abstract
Cancers of the central nervous system (CNS) are unique with respect to their tumor microenvironment. Such a status is due to immune-privilege and the cellular behaviors within a highly networked, neural-rich milieu. During tumor development in the CNS, neural, immune and cancer cells establish complex cell-to-cell communication networks which mimic physiological functions, including paracrine signaling and synapse-like formations. This crosstalk regulates diverse pathological functions contributing to tumor progression. In the CNS, regulation of physiological and pathological functions relies on various cell signaling and transcription programs. At the core of these events lies the cyclic adenosine monophosphate (cAMP) response element binding protein (CREB), a master transcriptional regulator in the CNS. CREB is a kinase inducible transcription factor which regulates many CNS functions, including neurogenesis, neuronal survival, neuronal activation and long-term memory. Here, we discuss how CREB-regulated mechanisms operating in diverse cell types, which control development and function of the CNS, are co-opted in CNS tumors.
Collapse
Affiliation(s)
- Marija Dinevska
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Samuel S Widodo
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Laura Cook
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Stanley S Stylli
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia; Department of Neurosurgery, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Robert G Ramsay
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology and the Department of Clinical Pathology, The University of Melbourne, Melbourne, Australia
| | - Theo Mantamadiotis
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia; Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia; Centre for Stem Cell Systems, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
47
|
Fontanella RA, Ghosh P, Pesapane A, Taktaz F, Puocci A, Franzese M, Feliciano MF, Tortorella G, Scisciola L, Sommella E, Ambrosino C, Paolisso G, Barbieri M. Tirzepatide prevents neurodegeneration through multiple molecular pathways. J Transl Med 2024; 22:114. [PMID: 38287296 PMCID: PMC10823712 DOI: 10.1186/s12967-024-04927-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 01/23/2024] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Several evidence demonstrated that glucagon-like peptide 1 receptor agonists (GLP1-RAs) reduce the risk of dementia in type 2 diabetes patients by improving memory, learning, and overcoming cognitive impairment. In this study, we elucidated the molecular processes underlying the protective effect of Tirzepatide (TIR), a dual glucose-dependent insulinotropic polypeptide receptor agonist (GIP-RA)/ GLP-1RA, against learning and memory disorders. METHODS We investigated the effects of TIR on markers of neuronal growth (CREB and BDNF), apoptosis (BAX/Bcl2 ratio) differentiation (pAkt, MAP2, GAP43, and AGBL4), and insulin resistance (GLUT1, GLUT4, GLUT3 and SORBS1) in a neuroblastoma cell line (SHSY5Y) exposed to normal and high glucose concentration. The potential role on DNA methylation of genes involved in neuroprotection and epigenetic modulators of neuronal growth (miRNA 34a), apoptosis (miRNA 212), and differentiation (miRNA 29c) was also investigated. The cell proliferation was detected by measuring Ki-67 through flow cytometry. The data were analysed by SPSS IBM Version 23 or GraphPad Prism 7.0 software and expressed as the means ± SEM. Differences between the mean values were considered significant at a p-value of < 0.05. GraphPad Prism software was used for drawing figures. RESULTS For the first time, it was highlighted: (a) the role of TIR in the activation of the pAkt/CREB/BDNF pathway and the downstream signaling cascade; (b) TIR efficacy in neuroprotection; (c) TIR counteracting of hyperglycemia and insulin resistance-related effects at the neuronal level. CONCLUSIONS We demonstrated that TIR can ameliorate high glucose-induced neurodegeneration and overcome neuronal insulin resistance. Thus, this study provides new insight into the potential role of TIR in improving diabetes-related neuropathy.
Collapse
Affiliation(s)
- Rosaria Anna Fontanella
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Puja Ghosh
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Ada Pesapane
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Fatemeh Taktaz
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Armando Puocci
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Martina Franzese
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Maria Federica Feliciano
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giovanni Tortorella
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Lucia Scisciola
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Eduardo Sommella
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy
| | - Concetta Ambrosino
- Biogem Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
- Department of Science and Technology, University of Sannio, Benevento, Italy
| | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
- UniCamillus, International Medical University, Rome, Italy
| | - Michelangela Barbieri
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
48
|
Atsumi Y, Iwata R, Kimura H, Vanderhaeghen P, Yamamoto N, Sugo N. Repetitive CREB-DNA interactions at gene loci predetermined by CBP induce activity-dependent gene expression in human cortical neurons. Cell Rep 2024; 43:113576. [PMID: 38128530 DOI: 10.1016/j.celrep.2023.113576] [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: 05/23/2023] [Revised: 11/10/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
Neuronal activity-dependent transcription plays a key role in plasticity and pathology in the brain. An intriguing question is how neuronal activity controls gene expression via interactions of transcription factors with DNA and chromatin modifiers in the nucleus. By utilizing single-molecule imaging in human embryonic stem cell (ESC)-derived cortical neurons, we demonstrate that neuronal activity increases repetitive emergence of cAMP response element-binding protein (CREB) at histone acetylation sites in the nucleus, where RNA polymerase II (RNAPII) accumulation and FOS expression occur rapidly. Neuronal activity also enhances co-localization of CREB and CREB-binding protein (CBP). Increased binding of a constitutively active CREB to CBP efficiently induces CREB repetitive emergence. On the other hand, the formation of histone acetylation sites is dependent on CBP histone modification via acetyltransferase (HAT) activity but is not affected by neuronal activity. Taken together, our results suggest that neuronal activity promotes repetitive CREB-CRE and CREB-CBP interactions at predetermined histone acetylation sites, leading to rapid gene expression.
Collapse
Affiliation(s)
- Yuri Atsumi
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Ryohei Iwata
- VIB-KU Leuven, Center for Brain & Disease Research and KU Leuven, Department of Neurosciences & Leuven Brain Institute, 3000 Leuven, Belgium
| | - Hiroshi Kimura
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Kanagawa 226-8503, Japan
| | - Pierre Vanderhaeghen
- VIB-KU Leuven, Center for Brain & Disease Research and KU Leuven, Department of Neurosciences & Leuven Brain Institute, 3000 Leuven, Belgium
| | - Nobuhiko Yamamoto
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan; Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518132, China.
| | - Noriyuki Sugo
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
49
|
Wan M, Sun S, Di X, Zhao M, Lu F, Zhang Z, Li Y. Icariin improves learning and memory function in Aβ 1-42-induced AD mice through regulation of the BDNF-TrκB signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:117029. [PMID: 37579923 DOI: 10.1016/j.jep.2023.117029] [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: 06/30/2023] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 08/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Epimedium brevicornu Maxim. is a traditional medicinal Chinese herb that is enriched with flavonoids, which have remarkably high medicinal value. Icariin (ICA) is a marker compound isolated from the total flavonoids of Epimedium brevicornu Maxim. It has been shown to improve Neurodegenerative disease, therefore, ICA is probably a potential drug for treating AD. MATERIALS AND METHODS The 6-8-week-old SPF-class male ICR mice were randomly divided into 8 groups for modeling, and then the mice were administered orally with ICA for 21 days. The behavioral experiments were conducted to evaluate if learning and memory behavior were absent in mice, confirming that infusion of Amyloid β-protein (Aβ)1-42 caused significant memory impairment. The morphological changes and damage of neurons in the mice's brains were observed by HE and Nissl staining. The spinous protrusions (dendritic spines) on neuronal dendrites were investigated by Golgi-Cox staining. The molecular mechanism of ICA was examined by Western Blot. The protein docking of ICA and Donepezil with BDNF were analyzed to determine their interaction. RESULTS The behavioral experimental results showed that in Aβ1-42-induced AD mice, the learning and memory abilities were improved after using ICA. At the same time, the low, medium, and high doses of ICA could reduce the content of Aβ1-42 in the hippocampus of AD mice, repair neuronal damage, enhance synaptic plasticity, as well as increase the expression of BDNF, TrκB, CREB, Akt, GAP43, PSD95, and SYN proteins in the hippocampus of mice. However, the effect with high doses of ICA is more pronounced. The high-dose administration of ICA has the best therapeutic effect on AD mice. After administering the inhibitor k252a, the therapeutic effect of ICA was reversed. The macromolecular docking results of ICA and BDNF protein demonstrated a strong interaction of -7.8 kcal/mol, which indicates that ICA plays a therapeutic role in AD mice by regulating the BDNF-TrκB signaling pathway. CONCLUSIONS The results confirm that ICA can repair neuronal damage, enhance synaptic plasticity, as well as ultimately improve learning and memory impairment through the regulation of the BDNF-TrκB signaling pathway.
Collapse
Affiliation(s)
- Meiyu Wan
- School of Pharmacy, North China University of Science and Technology, Tangshan, 063210, People's Republic of China
| | - Shengqi Sun
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, People's Republic of China
| | - Xiaoke Di
- School of Pharmacy, North China University of Science and Technology, Tangshan, 063210, People's Republic of China
| | - Minghui Zhao
- School of Pharmacy, North China University of Science and Technology, Tangshan, 063210, People's Republic of China
| | - Fengjuan Lu
- School of Pharmacy, North China University of Science and Technology, Tangshan, 063210, People's Republic of China
| | - Zhifei Zhang
- School of Pharmacy, North China University of Science and Technology, Tangshan, 063210, People's Republic of China
| | - Yang Li
- School of Pharmacy, North China University of Science and Technology, Tangshan, 063210, People's Republic of China.
| |
Collapse
|
50
|
Wang Y, Liang J, Xu B, Yang J, Wu Z, Cheng L. TrkB/BDNF signaling pathway and its small molecular agonists in CNS injury. Life Sci 2024; 336:122282. [PMID: 38008209 DOI: 10.1016/j.lfs.2023.122282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 10/19/2023] [Accepted: 11/18/2023] [Indexed: 11/28/2023]
Abstract
As one of the most prevalent neurotrophic factors in the central nervous system (CNS), brain-derived neurotrophic factor (BDNF) plays a significant role in CNS injury by binding to its specific receptor Tropomyosin-related kinase receptor B (TrkB). The BDNF/TrkB signaling pathway is crucial for neuronal survival, structural changes, and plasticity. BDNF acts as an axonal growth and extension factor, a pro-survival factor, and a synaptic modulator in the CNS. BDNF also plays an important role in the maintenance and plasticity of neuronal circuits. Several studies have demonstrated the importance of BDNF in the treatment and recovery of neurodegenerative and neurotraumatic disorders. By undertaking in-depth study on the mechanism of BDNF/TrkB function, important novel therapeutic strategies for treating neuropsychiatric disorders have been discovered. In this review, we discuss the expression patterns and mechanisms of the TrkB/BDNF signaling pathway in CNS damage and introduce several intriguing small molecule TrkB receptor agonists produced over the previous several decades.
Collapse
Affiliation(s)
- Yujin Wang
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; Medical School, Tongji University, Shanghai 200433, China
| | - Jing Liang
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; School of Stomatology, Tongji University, Shanghai 200072, China
| | - Boyu Xu
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; Medical School, Tongji University, Shanghai 200433, China
| | - Jin Yang
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; Medical School, Tongji University, Shanghai 200433, China
| | - Zhourui Wu
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China.
| | - Liming Cheng
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China.
| |
Collapse
|