1
|
Chen R, Chen W, Li P, Zhao Y, Zeng Q, Chen W, Cao D. Function and application of brain‑derived neurotrophic factor precursors (Review). Int J Mol Med 2025; 56:105. [PMID: 40341415 PMCID: PMC12081033 DOI: 10.3892/ijmm.2025.5546] [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: 01/26/2025] [Accepted: 04/29/2025] [Indexed: 05/10/2025] Open
Abstract
Brain‑derived neurotrophic factor precursor (proBDNF) plays a critical role in the pathogenesis and progression of various human diseases. Through its interaction with p75NTR and sortilin receptors, proBDNF promotes apoptosis, impairs synaptic plasticity, and contributes to the regulation of immune system function, inflammatory responses and cellular metabolic processes. proBDNF is widely distributed throughout the body, and as such, extensive research has demonstrated that proBDNF is significantly associated with the pathophysiological mechanisms underlying several diseases. In the present review, the mechanisms by which proBDNF contributes to different diseases are summarized to highlight its potential therapeutic and diagnostic implications. Specifically, the role of proBDNF in cognitive disorders, focusing on its effects on synaptic function and neural network dynamics, while analyzing the cascade reactions involving proBDNF and downstream effector molecules in inflammatory diseases, to elucidate its bidirectional regulatory effects in tumor initiation and progression. Furthermore, the function of proBDNF in neurogenesis, the mechanism by which it regulates the memory of fear, and enhances individual behavioral flexibility is discussed. Finally, the potential of proBDNF as a biomarker for disease diagnosis and the therapeutic prospects of targeting it using monoclonal antibodies are highlighted while also proposing future research directions. The present review can serve as a reference for translational medical research on proBDNF and its receptors.
Collapse
Affiliation(s)
- Risheng Chen
- Department of Anesthesiology, Pingshan Central Hospital of Shenzhen, Shenzhen, Guangdong 518122, P.R. China
| | - Weixin Chen
- Department of Science and Education, The Fourth People's Hospital of Shenzhen (Shenzhen Sami Medical Center), Shenzhen, Guangdong 518118, P.R. China
| | - Ping Li
- Department of Anesthesiology, Pingshan Central Hospital of Shenzhen, Shenzhen, Guangdong 518122, P.R. China
| | - Yingchang Zhao
- Department of Anesthesiology, Pingshan Central Hospital of Shenzhen, Shenzhen, Guangdong 518122, P.R. China
| | - Qianqian Zeng
- Department of Anesthesiology, Pingshan Central Hospital of Shenzhen, Shenzhen, Guangdong 518122, P.R. China
| | - Wenqing Chen
- Department of Anesthesiology, Pingshan Central Hospital of Shenzhen, Shenzhen, Guangdong 518122, P.R. China
| | - Dequan Cao
- Department of Anesthesiology, Pingshan Central Hospital of Shenzhen, Shenzhen, Guangdong 518122, P.R. China
| |
Collapse
|
2
|
Qiu Y, Zhu L, Cai W, Zhu L. Research Progress on BDNF and Depression. ACS Chem Neurosci 2025. [PMID: 40359301 DOI: 10.1021/acschemneuro.5c00193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025] Open
Abstract
Depression is a potentially life-threatening psychiatric disorder that affects the physical and mental health of millions of individuals worldwide. It can manifest at any stage of life, inducing profound emotional despondency, negative cognitions, and, in severe cases, suicidal ideation, often accompanied by physical symptoms, bringing a significant burden on both families and society. Brain-derived neurotrophic factor (BDNF), a member of the neurotrophic factor family, is widely expressed in the central nervous system (CNS), particularly in regions, such as the hippocampus and cortex. Numerous studies have shown that an imbalance or inadequate conversion of pro-brain-derived neurotrophic factor (proBDNF) into its mature form, mature BDNF (mBDNF), may impair neuronal plasticity, which is crucial to the pathogenesis of depression. This paper provides a comprehensive review of the neurotrophic mechanisms implicated in depression, covering the location, expression, and release of BDNF; the relationship between proBDNF, mBDNF, and depression; and the downstream signaling pathways triggered by BNDF binding to its receptors. This review aims to provide a theoretical foundation for understanding the pathogenesis and clinical treatment of depression.
Collapse
Affiliation(s)
- Yahong Qiu
- The Key Laboratory of Developmental Genes and Human Disease, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Lixia Zhu
- Patent Examination Cooperation (JIANGSU) Center of the Patent Office, China National Intellectual Property Administration (CNIPA), Suzhou, Jiangsu 215163, China
| | - Wenyan Cai
- The Key Laboratory of Developmental Genes and Human Disease, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Lijuan Zhu
- The Key Laboratory of Developmental Genes and Human Disease, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| |
Collapse
|
3
|
Li Y, Yang SY, Zhang YR, Wang Y. Decoding the neuroimmune axis in colorectal cancer: From neural circuitry to therapeutic innovation. Cytokine Growth Factor Rev 2025:S1359-6101(25)00044-9. [PMID: 40274426 DOI: 10.1016/j.cytogfr.2025.04.001] [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: 04/05/2025] [Revised: 04/15/2025] [Accepted: 04/15/2025] [Indexed: 04/26/2025]
Abstract
The nervous and immune systems are two major components that maintain body homeostasis, with their functional roles often overlapping significantly. Both systems are capable of identifying, integrating, and organizing responsive reactions to various external stimuli. The gut, referred to as the "second brain" and the largest immune organ in the body, serves as the most frequent focal site for neuroimmune interactions. Colorectal cancer (CRC), as the predominant solid tumor arising in this neuroimmune-rich microenvironment, remains understudied through the lens of neuroimmune regulatory mechanisms. This review systematically synthesizes current evidence to elucidate the neuroimmune axis in CRC pathogenesis, with particular emphasis on neuroimmune crosstalk-mediated remodeling of tumor immunity. We comprehensively catalog the immunomodulatory effects exerted by principal neuroregulatory mediators, categorized as: (1) neurotransmitters (glutamate, glutamine, γ-aminobutyric acid, epinephrine, norepinephrine, dopamine, serotonin, acetylcholine, and gaseous signaling molecules); (2) neuropeptides (substance P, calcitonin gene-related peptide, vasoactive intestinal peptide); and (3) neurotrophic factors. Furthermore, we critically evaluate the translational prospects and therapeutic challenges of targeting neuroimmune pathways and propose strategic priorities and research focuses for advancing the development of neuroimmune interaction-related therapeutic approaches in CRC.
Collapse
Affiliation(s)
- Ying Li
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Sheng-Ya Yang
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ying-Ru Zhang
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; The Second Clinical Medical College of Guizhou University of Traditional Chinese Medicine, Guizhou 550003, China.
| | - Yan Wang
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; The Second Clinical Medical College of Guizhou University of Traditional Chinese Medicine, Guizhou 550003, China.
| |
Collapse
|
4
|
Cholewa J, Nowacka-Chmielewska M, Gorzkowska A, Malecki A, Lasek-Bal A, Cholewa J. Changes in proBDNF and Mature BDNF Levels After Medium-Intensity Functional Motor Rehabilitation Program in Patients with Parkinson's Disease. Int J Mol Sci 2025; 26:3616. [PMID: 40332094 PMCID: PMC12027431 DOI: 10.3390/ijms26083616] [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: 01/25/2025] [Revised: 04/02/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
Physical rehabilitation complements the treatment of Parkinson's disease (PD). The applied physical exercises are effective in PD by promoting activity-dependent neuroplasticity. The main aim of this study was to assess the effect of a 16-week moderate-intensity functional physical rehabilitation program (FPR) on the concentration of mature brain-derived neurotrophic factor (BDNF) and its precursor (proBDNF) in blood serum and the severity of symptoms and quality of life in people with PD. People with PD (Hoehn and Yahr stage 3) were randomly assigned to the experimental (FPR) and control (CG) groups. FPR participated in movement training to improve functional mobility, motor coordination, and balance. Pre- and post-intervention assessments included serum levels of proBDNF, mature BDNF, MDS-UPDRS sub-scales, and the PDQ-39 quality of life measured. In the FPR group, a statistically significant increase in serum proBDNF levels by 39.42% (p = 0.006) was observed, as well as an improvement in motor and non-motor aspects of daily functioning, motor complications, and overall quality of life. No statistically significant changes in BDNF levels were observed. The results indicate that moderately intensive FPR enhances neurotrophic mechanisms, primarily through regulating proBDNF and improving motor functions and quality of life in patients with PD. The results underline the potential of targeted rehabilitation programs to increase neuroplasticity and improve clinical outcomes in PD.
Collapse
Affiliation(s)
- Joanna Cholewa
- Institute of Sport Sciences, Academy of Physical Education in Katowice, 40-065 Katowice, Poland;
| | - Marta Nowacka-Chmielewska
- Laboratory of Molecular Biology, Institute of Physiotherapy and Health Sciences, Academy of Physical Education in Katowice, 40-065 Katowice, Poland; (M.N.-C.); (A.M.)
| | - Agnieszka Gorzkowska
- Department of Neurology, School of Health Sciences, Medical University of Silesia in Katowice, 40-635 Katowice, Poland; (A.G.); (A.L.-B.)
| | - Andrzej Malecki
- Laboratory of Molecular Biology, Institute of Physiotherapy and Health Sciences, Academy of Physical Education in Katowice, 40-065 Katowice, Poland; (M.N.-C.); (A.M.)
| | - Anetta Lasek-Bal
- Department of Neurology, School of Health Sciences, Medical University of Silesia in Katowice, 40-635 Katowice, Poland; (A.G.); (A.L.-B.)
| | - Jaroslaw Cholewa
- Institute of Sport Sciences, Academy of Physical Education in Katowice, 40-065 Katowice, Poland;
| |
Collapse
|
5
|
Alnoaman H, Al-Kuraishy HM, Al-Gareeb AI, Turkistani A, Allam A, Alexiou A, Papadakis M, Batiha GES. Dysregulation of proBDNF/p75 NTR and BDNF/TrkB Signaling in Acute Ischemic Stroke: Different Sides of the Same Coins. Brain Res Bull 2025; 226:111338. [PMID: 40209946 DOI: 10.1016/j.brainresbull.2025.111338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 03/26/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
Acute ischemic stroke (AIS) is a focal neurological deficit due to sudden occlusion of cerebral vessels in the brain. AIS-induced neuronal injury and associated excite-toxicity and neurodegeneration affect the synthesis and the release of different neurotrophic factors such as brain-derived neurotropic factor (BDNF) and its precursor proBDNF. Both BDNF and proBDNF act on the specific receptors with different neurological effects. BDNF activates tropomyosin receptor kinase B (TrkB) receptor results in promoting neuronal survival, synaptic plasticity, and neuronal growth. However, the proBDNF activates p75 neurotrophin receptor (p75NTR) and sortilin which attenuates synaptic plasticity and promotes neuronal apoptosis. Dysregulation of central and peripheral expression of proBDNF/BDNF is linked with the severity and clinical outcomes of AIS. Therefore, this review aims to discuss the alterations of proBDNF/BDNF signaling in AIS. Findings from the present review illustrated that proBDNF/p75NTR/sortilin signaling pathway is exaggerated whereas; BDNF-TrkB signaling is reduced in AIS leading to neuronal apoptosis. Therefore, activation of BDNF-TrkB signaling, and inhibition of proBDNF/p75NTR/sortilin signaling pathway could be a promising therapeutic strategy in the management of AIS.
Collapse
Affiliation(s)
- Hala Alnoaman
- Consultant family medicine, Ministry of health, Kingdom of Saudi Arabia.
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad 14132, Iraq.
| | - Ali I Al-Gareeb
- Head of Jabir Ibn, Hayyan Medical University, Al-Ameer Qu./Najaf-Iraq, Kufa PO.Box13, Iraq,.
| | - Areej Turkistani
- Department of pharmacology and toxicology, Collage of Medicine, Taif University,Taif 21944, Kingdom of Saudi Arabia.
| | - Albatoul Allam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), AL-Azhar University, Cairo, Egypt.
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Mohali, India; Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, New South Wales, Australia; Department of Research & Development, Funogen, Athens, Greece; Department of Research & Development, AFNP Med, Wien, Austria.
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, Wuppertal, 42283, Germany.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt.
| |
Collapse
|
6
|
Wang L, Su F, Huang H, Jiang Q, Kong H, Pei Z. Application of mRNA technology in neuronal protection of human mature brain-derived neurotrophic factor. Tissue Cell 2025; 93:102788. [PMID: 39933411 DOI: 10.1016/j.tice.2025.102788] [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: 06/06/2024] [Revised: 02/02/2025] [Accepted: 02/06/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND Although human mature brain-derived neurotrophic factor (hmBDNF) offers potential neuronal protection, its clinical translation remains challenging. Messenger RNA (mRNA) technology is promising in selectively upregulating protein cleavage products. This proof-of-concept study aims to evaluate the neuronal protective effects of hmBDNF mRNA in vitro. METHODS We optimized and synthesized hmBDNF mRNA and conducted dose-response and time-response analyses in SH-SY5Y cells. mRNA expression was assessed via qPCR, while protein expression was evaluated through immunostaining and ELISA. Cell survival rate was measured using cell counting kit-8. We examined cell survival rates in both differentiated and non-differentiated SH-SY5Y cells exposed to H2O2 or serum deprivation following hmBDNF mRNA incubation. Additionally, we assessed the expression of synapse-relevant genes (MAP2, synaptophysin) and the mBDNF receptor (TrkB) in both cell types. RESULTS The optimized hmBDNF mRNA effectively upregulated hmBDNF expression in SH-SY5Y cells with minimal impact on endogenous proBDNF expression. Dose-response and time-response analyses identified the optimal dose and time point for maximum hmBDNF expression. hmBDNF mRNA significantly increased cell survival in differentiated SH-SY5Y cells expressing MAP2, synaptophysin and TrkB after exposure to oxidative stress or serum deprivation. However, hmBDNF mRNA did not enhance cell survival in non-differentiated SH-SY5Y cells. CONCLUSION The optimized hmBDNF mRNA demonstrated a capacity for neuronal protection in vitro. Further in-vivo studies are required to assess its potential for clinical translation.
Collapse
Affiliation(s)
- Liang Wang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, Guangzhou 510080, China; National Key Clinical Department and Key Discipline of Neurology, Guangzhou 510080, China
| | - Fengjuan Su
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, Guangzhou 510080, China; National Key Clinical Department and Key Discipline of Neurology, Guangzhou 510080, China
| | - Heng Huang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, Guangzhou 510080, China; National Key Clinical Department and Key Discipline of Neurology, Guangzhou 510080, China
| | - Qiuhong Jiang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, Guangzhou 510080, China; National Key Clinical Department and Key Discipline of Neurology, Guangzhou 510080, China
| | - Haifang Kong
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, Guangzhou 510080, China; National Key Clinical Department and Key Discipline of Neurology, Guangzhou 510080, China
| | - Zhong Pei
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, Guangzhou 510080, China; National Key Clinical Department and Key Discipline of Neurology, Guangzhou 510080, China.
| |
Collapse
|
7
|
Edman S, Starck J, Corell L, Hangasjärvi W, von Finckenstein A, Reimeringer M, Reitzner S, Norrbom J, Moberg M, von Walden F. Exercise-induced plasma mature brain-derived neurotrophic factor elevation in children, adolescents and adults: influence of age, maturity and physical activity. J Physiol 2025; 603:2333-2347. [PMID: 40167390 PMCID: PMC12013800 DOI: 10.1113/jp288170] [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: 11/22/2024] [Accepted: 03/04/2025] [Indexed: 04/02/2025] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a neurotrophin that plays a central role in neuronal health. BDNF exists in two primary isoforms, the mature form (mBDNF) and its precursor (proBDNF), with opposing downstream effects on neuronal function. The positive effect of exercise on plasma levels of the BDNF isoforms has been extensively studied in adults. However, equivalent investigations are lacking in children and adolescents. Twenty healthy children (9-12 years old), 19 adolescents (13-17 years old) and 39 adults (23-49 years old) donated venous blood before and after a 45-minute run. Platelet-poor plasma was analysed for pro- and mBDNF using an enzyme-linked immunosorbent assay. Maximal oxygen uptake and anthropometric data were assessed in all participants, while Tanner stage, circulating sex hormones and accelerometry-based activity level were assessed in children and adolescents only. We found that children, adolescents and adults have similar circulating levels of plasma pro- and mBDNF at rest. For children and adolescents, resting levels of mBDNF correlated with average time spent in vigorous activity. In response to the acute endurance exercise intervention, mBDNF increased in all age groups, but the greatest rise in mBDNF was seen in adults. The acute endurance exercise did not affect proBDNF levels. Our results demonstrate that plasma mBDNF levels, but not proBDNF, increase following endurance exercise in all age groups, with a greater effect in adults. We also show that high-intensity physical activity, but not underlying fitness, is contributing to sustained elevated mBDNF levels. KEY POINTS: We show that in children and adolescents, regular vigorous physical activity is key to increased basal levels of plasma mature brain-derived neurotrophic factor (mBDNF), a factor linked to neuroplasticity and brain health. The ability to elevate mBDNF through exercise is present across all age groups, with the greatest increase in adults. The mBDNF response to physical exercise seems to be independent of underlying physical fitness. Our findings suggest that basal plasma mBDNF levels may reflect the cumulative effects of repeated exercise rather than an individual's overall physical fitness.
Collapse
Affiliation(s)
- Sebastian Edman
- Department of Women's and Children's HealthKarolinska InstituteStockholmSweden
- Department of Physiology and PharmacologyKarolinska InstituteStockholmSweden
| | - Julia Starck
- Department of Women's and Children's HealthKarolinska InstituteStockholmSweden
| | - Linnéa Corell
- Department of Women's and Children's HealthKarolinska InstituteStockholmSweden
| | - William Hangasjärvi
- Department of Physiology and PharmacologyKarolinska InstituteStockholmSweden
| | | | - Mikael Reimeringer
- Department of Women's and Children's HealthKarolinska InstituteStockholmSweden
| | - Stefan Reitzner
- Department of Women's and Children's HealthKarolinska InstituteStockholmSweden
- Department of Physiology and PharmacologyKarolinska InstituteStockholmSweden
| | - Jessica Norrbom
- Department of Physiology and PharmacologyKarolinska InstituteStockholmSweden
| | - Marcus Moberg
- Department of Physiology and PharmacologyKarolinska InstituteStockholmSweden
- Department of Physiology, Nutrition, and BiomechanicsThe Swedish School of Sport and Health SciencesStockholmSweden
| | | |
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
|
Barakat N, Brunelin J, Abrial E, Neige C, Chalancon B, Scholl J, Fourier A, Brossaud J, Hermand M, Besch V, Simon L, Magnin C, Leaune E, Poulet E. Suicide reattempts in adolescents and young adults after a first suicide attempt. Results from the SURAYA prospective cohort study. L'ENCEPHALE 2025:S0013-7006(25)00030-2. [PMID: 40089438 DOI: 10.1016/j.encep.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/11/2024] [Accepted: 12/18/2024] [Indexed: 03/17/2025]
Abstract
INTRODUCTION Suicide is a particularly major public health concern among young people as it is the fourth cause of death in youth aged 15-29. Despite various prevention strategies and efforts, the rate of suicide attempts among youth has increased over time and has risen even further since the COVID-19 pandemic. OBJECTIVES The aim of this study was to ascertain the prevalence of suicide reattempts within 3months in a cohort of young first-time suicide attempters aged 16 to 25years. Exploratory objectives were to investigate potential risk factors associated with reattempts in this population. METHODS We conducted a prospective, naturalistic, single-center cohort study including 182 patients hospitalized in emergency psychiatry for a first suicide attempt. Data on 31 sociodemographic, clinical and biological factors known to be associated with suicide were collected at baseline. RESULTS Out of the 182 patients included, 146 participants remained for the 3-months follow-up analysis (mean age: 19.71±2.5years, 71.9% female). Twenty of them reattempted suicide: yielding a prevalence of 13.7% (14.3% of females and 12.2% of males). Only four clinical and biological factors under study were significantly associated with suicide reattempt. CONCLUSION Our findings underscore the critical need for targeted prevention strategies for adolescents and young adults, as they represent a high-risk group for early suicide reattempts. Further research into the factors associated with recurrent suicide attempts is essential to more accurately characterize the profiles of young individuals who reattempt suicide, thereby informing the development of effective preventive interventions and avoiding negative outcomes.
Collapse
Affiliation(s)
- Nadine Barakat
- Le Vinatier, Psychiatrie Universitaire Lyon Métropole, 69500 Bron, France; Université Claude-Bernard Lyon 1, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre de Recherche en Neurosciences de Lyon U1028 UMR5292, PSYR2, 69500 Bron, France
| | - Jérôme Brunelin
- Le Vinatier, Psychiatrie Universitaire Lyon Métropole, 69500 Bron, France; Université Claude-Bernard Lyon 1, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre de Recherche en Neurosciences de Lyon U1028 UMR5292, PSYR2, 69500 Bron, France
| | - Erika Abrial
- Université Claude-Bernard Lyon 1, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre de Recherche en Neurosciences de Lyon U1028 UMR5292, PSYR2, 69500 Bron, France; University Hospital Édouard-Herriot, Hospices Civils de Lyon, 69000 Lyon, France
| | - Cécilia Neige
- Le Vinatier, Psychiatrie Universitaire Lyon Métropole, 69500 Bron, France; Université Claude-Bernard Lyon 1, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre de Recherche en Neurosciences de Lyon U1028 UMR5292, PSYR2, 69500 Bron, France
| | - Benoît Chalancon
- Le Vinatier, Psychiatrie Universitaire Lyon Métropole, 69500 Bron, France
| | - Jacqueline Scholl
- Le Vinatier, Psychiatrie Universitaire Lyon Métropole, 69500 Bron, France; Université Claude-Bernard Lyon 1, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre de Recherche en Neurosciences de Lyon U1028 UMR5292, PSYR2, 69500 Bron, France
| | - Anthony Fourier
- Laboratory of Medical Biology and Anatomo-Pathology, Hospices Civils de Lyon, Groupement Hospitalier Est, 69500 Bron, France
| | - Julie Brossaud
- University of Bordeaux, NutriNeurO, UMR 1286, CHU de Bordeaux, Laboratory of hormonology, Bordeaux, France
| | - Marianne Hermand
- Louis-Mourier Hospital, Assistance publique-Hôpitaux de Paris, 92700 Colombes, France
| | - Vincent Besch
- Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland; University of Geneva, Geneva, Switzerland
| | - Louis Simon
- University Hospital Édouard-Herriot, Hospices Civils de Lyon, 69000 Lyon, France
| | - Charline Magnin
- University Hospital Édouard-Herriot, Hospices Civils de Lyon, 69000 Lyon, France
| | - Edouard Leaune
- Le Vinatier, Psychiatrie Universitaire Lyon Métropole, 69500 Bron, France; Université Claude-Bernard Lyon 1, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre de Recherche en Neurosciences de Lyon U1028 UMR5292, PSYR2, 69500 Bron, France; Université Claude-Bernard Lyon 1, Institut National de la Santé et de la Recherche Médicale U1290, Lyon, France
| | - Emmanuel Poulet
- Le Vinatier, Psychiatrie Universitaire Lyon Métropole, 69500 Bron, France; Université Claude-Bernard Lyon 1, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre de Recherche en Neurosciences de Lyon U1028 UMR5292, PSYR2, 69500 Bron, France; University Hospital Édouard-Herriot, Hospices Civils de Lyon, 69000 Lyon, France.
| |
Collapse
|
10
|
Jenwitheesuk A, Pabalan N, Tapanadechopone P, Jarjanazi H, Arunphalungsanti K, Tharabenjasin P. Association of Brain-derived Neurotrophic Factor Polymorphisms With Alcohol Use Disorder: An Updated Meta-Analysis of Genetic Association Studies. Brain Behav 2025; 15:e70359. [PMID: 40021949 PMCID: PMC11870837 DOI: 10.1002/brb3.70359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 12/14/2024] [Accepted: 01/10/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) has been proposed to play a role in chronic alcohol consumption. However, studies investigating the association of single nucleotide polymorphisms (SNPs) in the BDNF gene with alcohol use disorder (AUD), including alcohol dependence, have obtained inconsistent results. This meta-analysis aims to examine the role of BDNF SNPs (rs6265, rs16917204, rs7103411, and rs11030104) in the risk of AUD. MATERIALS AND METHODS A multidatabase search identified 17 articles (20 studies) for inclusion. Pooled odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to estimate associations using standard genetic models (homozygous, recessive, dominant, and codominant). Significant associations were defined as those with a p-value ≤ 0.05 after applying the Bonferroni correction (pBC). Subgroup analysis was conducted based on ethnicity (Caucasian and Asian populations). Sources of heterogeneity were investigated through outlier treatment and meta-regression analysis. Only significant outcomes were further subjected to sensitivity analysis and assessment of publication bias. RESULTS This meta-analysis generated four significant pooled ORs, representing the core outcomes, all of which indicated reduced risks. Overall, the results indicated a significant association between the BDNF polymorphism and the risk of AUD in homozygous (OR = 0.72, 95% CIs = 0.60-0.85, pBC = 0.0038) and codominant (OR = 0.84, 95% CIs = 0.78-0.91, pBC = 0.0019) model. In subgroup analysis by ethnicity, homozygous (OR = 0.59, 95% CIs = 0.44-0.78, pBC = 0.0057) and recessive (OR = 0.61, 95% CIs = 0.46-0.81, pBC = 0.0133) models of BDNF polymorphisms were significantly associated with a reduced risk of AUD in Caucasians. However, no significant associations were found in Asians. Meta-regression analysis did not identify any covariates that significantly contributed to the observed heterogeneity. The core significant associations were robust and showed no evidence of publication bias. CONCLUSION The current meta-analysis suggests that the examined BDNF SNPs have a protective effect in the overall analysis (homozygous and codominant) and in the Caucasians subgroup (homozygous and recessive) while the Asians exhibited no effects of BDNF SNPs on AUD. BDNF polymorphisms might serve as a protective factor against the risk of AUD and could be useful markers in the clinical genetics of AUD.
Collapse
Affiliation(s)
- Anorut Jenwitheesuk
- Princess Agrarajakumari College of NursingChulabhorn Royal AcademyBangkokThailand
| | - Noel Pabalan
- Chulabhorn International College of MedicineThammasat UniversityPathum ThaniThailand
| | | | - Hamdi Jarjanazi
- Environmental Monitoring and Reporting Branch, Ontario Ministry of the Environment, Conservation and ParksTorontoOntarioCanada
| | | | | |
Collapse
|
11
|
Bogacheva PO, Potapova DA, Gaydukov AE. Sortilin and L-type Calcium Channels May be Involved in the Unusual Mechanism of proBDNF Signaling in Regenerating Mouse Neuromuscular Junctions. Neurochem Res 2025; 50:104. [PMID: 39998597 DOI: 10.1007/s11064-025-04360-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/17/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025]
Abstract
proBDNF and its main proteolytic product BDNF play crucial roles in maturation of neuromuscular junctions during development or reinnervation. We investigated the mechanisms of acute proBDNF effects on synaptic transmission in mouse motor synapses regenerating after nerve crush. The cleavage-resistant proBDNF mimicked the previously shown effect of cleavable proBDNF- GIRK-mediated decrease in the miniature endplate potential (MEPP) frequency accompanied by slight hyperpolarization of postsynaptic membrane. Remarkably, this effect did not utilize canonical proBDNF signaling pathway since inhibition of either p75 receptors with LM11A-31 or sortilin with AF38469 was not able to prevent it. Without sortilin activity, proBDNF downregulated the quantal content of multiquantal endplate potentials (EPP). This non-canonical action of proneurotrophin via TrkB receptors highlights the important role of sortilin as a safeguard preventing the spread of the negative effect of proBDNF on the evoked neurotransmitter release in regenerating motor synapses. In the absence of sortilin activity L-type calcium channels emerged as the key players providing proBDNF-induced decrease of EPP quantal content, while they were not involved in proBDNF-induced decrease of MEPP frequency. Sortilin-independent but TrkB- and GIRK-mediated inhibition of spontaneous release by proBDNF was not associated with the activity of acetylcholine (M2) or purinergic (A1 and P2Y13) metabotropic receptors. We propose that depending on sortilin involvement, proBDNF selectively affects spontaneous or evoked quantal neurotransmitter release via different branches of signaling pathway that ensure the presynaptic activation of GIRK or L-type calcium channels, respectively.
Collapse
Affiliation(s)
- P O Bogacheva
- Faculty of Biology, Department of Human and Animal Physiology, Lomonosov Moscow State University, Moscow, Russian Federation.
| | - D A Potapova
- Faculty of Biology, Department of Human and Animal Physiology, Lomonosov Moscow State University, Moscow, Russian Federation
| | - A E Gaydukov
- Faculty of Biology, Department of Human and Animal Physiology, Lomonosov Moscow State University, Moscow, Russian Federation
| |
Collapse
|
12
|
Numakawa T, Kajihara R. The Role of Brain-Derived Neurotrophic Factor as an Essential Mediator in Neuronal Functions and the Therapeutic Potential of Its Mimetics for Neuroprotection in Neurologic and Psychiatric Disorders. Molecules 2025; 30:848. [PMID: 40005159 PMCID: PMC11857940 DOI: 10.3390/molecules30040848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/04/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Among neurotrophins, including nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), and neurotrophin-4 (NT-4/5), BDNF has been extensively studied for its physiological role in cell survival and synaptic regulation in the central nervous system's (CNS's) neurons. BDNF binds to TrkB (a tyrosine kinase) with high affinity, and the resulting downstream intracellular signaling cascades play crucial roles in determining cell fate, including neuronal differentiation and maturation of the CNS neurons. It has been well demonstrated that the downregulation/dysregulation of the BDNF/TrkB system is implicated in the pathogenesis of neurologic and psychiatric disorders, such as Alzheimer's disease (AD) and depression. Interestingly, the effects of BDNF mimetic compounds including flavonoids, small molecules which can activate TrkB-mediated signaling, have been extensively investigated as potential therapeutic strategies for brain diseases, given that p75NTR, a common neurotrophin receptor, also contributes to cell death under a variety of pathological conditions such as neurodegeneration. Since the downregulation of the BDNF/TrkB system is associated with the pathophysiology of neurodegenerative diseases and psychiatric disorders, understanding how alterations in the BDNF/TrkB system contribute to disease progression could provide valuable insight for the prevention of these brain diseases. The present review shows recent advances in the molecular mechanisms underlying the BDNF/TrkB system in neuronal survival and plasticity, providing critical insights into the potential therapeutic impact of BDNF mimetics in the pathophysiology of brain diseases.
Collapse
Affiliation(s)
- Tadahiro Numakawa
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Ryutaro Kajihara
- Department of Hematology and Immunology, Faculty of Life Science, Kumamoto University, Kumamoto 862-0976, Japan
| |
Collapse
|
13
|
Gusar V, Kan N, Leonova A, Chagovets V, Tyutyunnik V, Khachatryan Z, Yarotskaya E, Sukhikh G. Non-Invasive Assessment of Neurogenesis Dysfunction in Fetuses with Early-Onset Growth Restriction Using Fetal Neuronal Exosomes Isolating from Maternal Blood: A Pilot Study. Int J Mol Sci 2025; 26:1497. [PMID: 40003962 PMCID: PMC11855093 DOI: 10.3390/ijms26041497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
The vector of modern obstetrics is aimed at finding ways to predict various placenta-associated complications, including those associated with neuronal dysfunction on in fetal growth restriction (FGR). The technology of fetal neuronal exosome (FNE) isolation from the maternal bloodstream opens up unique opportunities for detecting early signs of fetal brain damage. Using this method, FNEs were isolated from the blood of pregnant women with and without early-onset FGR, and the expression of a number of proteins in their composition was assessed (Western blotting). Significant changes in the level of proteins involved in neurogenesis (pro-BDNF (brain-derived neurotrophic factor), pro-NGF (nerve growth factor), TAG1/Contactin2) and presynaptic transmission (Synapsin 1, Synaptophysin) were revealed. The preliminary data on the expression of FNE proteins that perform post-translational modifications-sumoylation (SUMO 1, UBC9) and neddylation (NEDD8, UBC12)-were obtained. A relationship was established between altered protein expression and neonatal outcomes in newborns with growth restriction. Our study opens up new possibilities for non-invasive prenatal monitoring of fetal neurodevelopment disorders and possibilities of their correction in placenta-associated diseases.
Collapse
Affiliation(s)
- Vladislava Gusar
- Laboratory of Applied Transcriptomics, Federal State Budget Institution “National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov” of the Ministry of Health of the Russian Federation, 4, Oparina Street, 117997 Moscow, Russia
| | - Natalia Kan
- Federal State Budget Institution “National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov” of the Ministry of Health of the Russian Federation, 4, Oparina Street, 117997 Moscow, Russia; (N.K.); (E.Y.); (G.S.)
| | - Anastasia Leonova
- Department of Molecular Diagnostic Methods and Personalized Medicine, Federal State Budget Institution “National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov” of the Ministry of Health of the Russian Federation, 4, Oparina Street, 117997 Moscow, Russia;
| | - Vitaliy Chagovets
- Laboratory of Metabolomics and Bioinformatics, Federal State Budget Institution “National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov” of the Ministry of Health of the Russian Federation, 4, Oparina Street, 117997 Moscow, Russia;
| | - Victor Tyutyunnik
- Center for Scientific and Clinical Research, Federal State Budget Institution “National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov” of the Ministry of Health of the Russian Federation, 4, Oparina Street, 117997 Moscow, Russia;
| | - Zarine Khachatryan
- JSC “European Medical Center”, 35, Shchepkina Street, 129090 Moscow, Russia;
| | - Ekaterina Yarotskaya
- Federal State Budget Institution “National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov” of the Ministry of Health of the Russian Federation, 4, Oparina Street, 117997 Moscow, Russia; (N.K.); (E.Y.); (G.S.)
| | - Gennadiy Sukhikh
- Federal State Budget Institution “National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov” of the Ministry of Health of the Russian Federation, 4, Oparina Street, 117997 Moscow, Russia; (N.K.); (E.Y.); (G.S.)
| |
Collapse
|
14
|
Carrasco M, Guzman L, Olloquequi J, Cano A, Fortuna A, Vazquez-Carrera M, Verdaguer E, Auladell C, Ettcheto M, Camins A. Licochalcone A prevents cognitive decline in a lipopolysaccharide-induced neuroinflammation mice model. Mol Med 2025; 31:54. [PMID: 39930360 PMCID: PMC11812219 DOI: 10.1186/s10020-025-01106-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 01/27/2025] [Indexed: 02/13/2025] Open
Abstract
Inflammation plays a key role in the development of neurodegenerative disorders that are currently incurable. Licochalcone A (LCA) has been described as an emerging anti-inflammatory drug with multiple therapeutical properties that could potentially prevent neurodegeneration. However, its neuroprotective mechanism remains unclear. Here, we investigated if LCA prevents cognitive decline induced by Lipopolysaccharide (LPS) and elucidated its potential benefits. For that, 8-week-old C57BL6/J male mice were intraperitonially (i.p.) treated with saline solution or LCA (15 mg/kg/day, 3 times per week) for two weeks. The last day, a single i.p injection of LPS (1 mg/kg) or saline solution was administered 24 h before sacrifice. The results revealed a significant reduction in mRNA expression in genes involved in oxidative stress (Sod1, Cat, Pkm, Pdha1, Ndyfv1, Uqcrb1, Cycs and Cox4i1), metabolism (Slc2a1, Slc2a2, Prkaa1 and Gsk3b) and synapsis (Bdnf, Nrxn3 and Nlgn2) in LPS group compared to saline. These findings were linked to memory impairment and depressive-like behavior observed in this group. Interestingly, LCA protected against LPS alterations through its anti-inflammatory effect, reducing gliosis and regulating M1/M2 markers. Moreover, LCA-treated animals showed a significant improvement of antioxidant mechanisms, such as citrate synthase activity and SOD2. Additionally, LCA demonstrated protection against metabolic disturbances, downregulating GLUT4 and P-AKT, and enhanced the expression of synaptic-related proteins (P-CREB, BDNF, PSD95, DBN1 and NLG3), leading all together to dendritic spine preservation. In conclusion, our results demonstrate that LCA treatment prevents LPS-induced cognitive decline by reducing inflammation, enhancing the antioxidant response, protecting against metabolic disruptions and improving synapsis related mechanisms.
Collapse
Affiliation(s)
- Marina Carrasco
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Laura Guzman
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
| | - Jordi Olloquequi
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Talca, Chile
| | - Amanda Cano
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Ana Fortuna
- Laboratory of Pharmacology and Pharmaceutical Care, Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research, CIBIT/ICNAS, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Manuel Vazquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain
- Networking Research Centre of Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, 28031, Madrid, Spain
- Institute of Biomedicine of the Universitat de Barcelona (IBUB), University of Barcelona, 08028, Barcelona, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, 08950, Esplugues de Llobregat, Spain
| | - Ester Verdaguer
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028, Barcelona, Spain
| | - Carme Auladell
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028, Barcelona, Spain
| | - Miren Ettcheto
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain.
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain.
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain.
| | - Antoni Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| |
Collapse
|
15
|
Siebert JR, Kennedy K, Osterhout DJ. Neurons Are Not All the Same: Diversity in Neuronal Populations and Their Intrinsic Responses to Spinal Cord Injury. ASN Neuro 2025; 17:2440299. [PMID: 39819292 PMCID: PMC11877619 DOI: 10.1080/17590914.2024.2440299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025] Open
Abstract
Functional recovery following spinal cord injury will require the regeneration and repair of damaged neuronal pathways. It is well known that the tissue response to injury involves inflammation and the formation of a glial scar at the lesion site, which significantly impairs the capacity for neuronal regeneration and functional recovery. There are initial attempts by both supraspinal and intraspinal neurons to regenerate damaged axons, often influenced by the neighboring tissue pathology. Many experimental therapeutic strategies are targeted to further stimulate the initial axonal regrowth, with little consideration for the diversity of the affected neuronal populations. Notably, recent studies reveal that the neuronal response to injury is variable, based on multiple factors, including the location of the injury with respect to the neuronal cell bodies and the affected neuronal populations. New insights into regenerative mechanisms have shown that neurons are not homogenous but instead exhibit a wide array of diversity in their gene expression, physiology, and intrinsic responses to injury. Understanding this diverse intrinsic response is crucial, as complete functional recovery requires the successful coordinated regeneration and reorganization of various neuron pathways.
Collapse
Affiliation(s)
- Justin R. Siebert
- Physician Assistant Studies Program, Department of Health Care and Administration, Slippery Rock University of Pennsylvania, Slippery Rock, PA, USA
| | - Kiersten Kennedy
- Norton College of Medicine, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Donna J. Osterhout
- Department of Cell & Developmental Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
16
|
Chen B, Sun Y, Sun H, Cong N, Ma R, Qian X, Lyu J, Fu X, Chi F, Li H, Liu Y, Ren D, Bu W. Ultrasound-Triggered NO Release to Promote Axonal Regeneration for Noise-Induced Hearing Loss Therapy. ACS NANO 2024; 18:33232-33244. [PMID: 39561026 DOI: 10.1021/acsnano.4c12676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
Intense noise poses a threat to spiral ganglion neurons (SGNs) in the inner ear, often resulting in limited axonal regeneration during noise injury and leading to noise-induced hearing loss (NIHL). Here, we propose an ultrasound-triggered nitric oxide (NO) release to enhance the sprouting and regeneration of injured axons in SGNs. We developed hollow silicon nanoparticles to load nitrosylated N-acetylcysteine, producing HMSN-SNO, which effectively protects NO from external interferences. Utilizing low-intensity ultrasound stimulation with bone penetration, we achieve the controlled release of NO from HMSN-SNO within the cochlea. In mice with NIHL, a rapid and extensive loss of synaptic connections between hair cells and SGNs is observed within 24 h after exposure to excessive noise. However, this loss could be reversed with the combined treatment, resulting in a hearing functional recovery from 83.57 to 65.00 dB SPL. This positive outcome is attributed to the multifunctional effects of HMSN-SNO, wherein they scavenge reactive oxygen species (ROS) to reverse the pathological microenvironment and simultaneously upregulate the CREB/BDNF/EGR1 signaling pathway, thereby enhancing neuroplasticity and promoting the regeneration of neuronal axons. These findings underscore the potential of nanomedicine for neuroplasticity modulation, which holds promise for advancing both basic research and the further treatment of neurological diseases.
Collapse
Affiliation(s)
- Binjun Chen
- ENT institute and Department of Otorhinolaryngology, NHC Key Laboratory of Hearing Medicine. Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai 200031, China
| | - Yanhong Sun
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Academy for Engineering and Technology, Fudan University, 220 Handan Road, Shanghai 200438, China
| | - Haojie Sun
- ENT institute and Department of Otorhinolaryngology, NHC Key Laboratory of Hearing Medicine. Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai 200031, China
| | - Ning Cong
- ENT institute and Department of Otorhinolaryngology, NHC Key Laboratory of Hearing Medicine. Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai 200031, China
| | - Rui Ma
- ENT institute and Department of Otorhinolaryngology, NHC Key Laboratory of Hearing Medicine. Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai 200031, China
| | - Xiaoqing Qian
- ENT institute and Department of Otorhinolaryngology, NHC Key Laboratory of Hearing Medicine. Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai 200031, China
| | - Jihan Lyu
- ENT institute and Department of Otorhinolaryngology, NHC Key Laboratory of Hearing Medicine. Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai 200031, China
| | - Xiao Fu
- ENT institute and Department of Otorhinolaryngology, NHC Key Laboratory of Hearing Medicine. Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai 200031, China
| | - Fanglu Chi
- ENT institute and Department of Otorhinolaryngology, NHC Key Laboratory of Hearing Medicine. Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai 200031, China
| | - Hongzhe Li
- Research Service, VA Loma Linda Healthcare System, 11201 Benton Street, Loma Linda, CA 92357, USA
| | - Yanyan Liu
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Academy for Engineering and Technology, Fudan University, 220 Handan Road, Shanghai 200438, China
| | - Dongdong Ren
- ENT institute and Department of Otorhinolaryngology, NHC Key Laboratory of Hearing Medicine. Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai 200031, China
| | - Wenbo Bu
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Academy for Engineering and Technology, Fudan University, 220 Handan Road, Shanghai 200438, China
| |
Collapse
|
17
|
Henderson J, O'Callaghan J, Campbell M. Gene therapy for glaucoma: Targeting key mechanisms. Vision Res 2024; 225:108502. [PMID: 39423611 PMCID: PMC11579448 DOI: 10.1016/j.visres.2024.108502] [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: 07/31/2024] [Revised: 10/04/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024]
Abstract
Glaucoma is a group of optic neuropathies characterised by progressive retinal ganglion cell (RGC) degeneration and is the leading cause of irreversible blindness worldwide. Current treatments for glaucoma focus on reducing intraocular pressure (IOP) with topical medications. However, many patients do not achieve sufficient IOP reductions with such treatments. Patient compliance to dosing schedules also poses a significant challenge, further limiting their effectiveness. While surgical options exist for resistant cases, these are invasive and carry risks of complications. Thus, there is a critical need for better strategies to prevent irreversible vision loss in glaucoma. Gene therapy holds significant promise in this regard, offering potential long-term solutions by targeting the disease's underlying causes at a molecular level. Gene therapy strategies for glaucoma primarily target the two key hallmarks of the disease: elevated IOP and RGC death. This review explores key mechanisms underlying these hallmarks and discusses the current state of gene therapies targeting them. In terms of IOP reduction, this review covers strategies aimed at enhancing extracellular matrix turnover in the conventional outflow pathway, targeting fibrosis, regulating aqueous humor production, and targeting myocilin for gene-specific therapy. Neuroprotective strategies explored include targeting neurotrophic factors and their receptors, reducing oxidative stress and mitochondrial dysfunction, and preventing Wallerian degeneration. This review also briefly highlights key research priorities for advancing gene therapies for glaucoma through the clinical pipeline, such as refining delivery vectors and improving transgene regulation. Addressing these priorities will be essential for translating advancements from preclinical models into effective clinical therapies for glaucoma.
Collapse
Affiliation(s)
- Jeff Henderson
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | | | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
18
|
Peng YY, Tang C, Wang HY, Ding Y, Yang H, Ma XM, Gao J, Li S, Long ZY, Lu XM, Wang YT. p75NTR mediated chronic restraint stress-induced depression-like behaviors in mice via hippocampal mTOR pathway. Life Sci 2024; 358:123175. [PMID: 39477145 DOI: 10.1016/j.lfs.2024.123175] [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: 07/20/2024] [Revised: 10/01/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024]
Abstract
AIMS Major depressive disorder (MDD) is an enduring and severe mood disorder. Previous studies have indicated that p75NTR is involved in neuronal survival and death. However, the specific mechanism of p75NTR in depression remains unknown. The present study aimed to explore the role and mechanism of p75NTR in depression, and try to provide a new target for the treatment of MDD. MAIN METHODS The p75NTR knockout and overexpression mice were used to establish a mouse model of depression induced by chronic restraint stress (CRS), and the behavioral effects and potential mechanisms associated with p75NTR knockout/overexpression on CRS-induced depressive mice were investigated by animal behavior, histopathology, immunofluorescence and western blot, respectively. KEY FINDINGS The results demonstrate that p75NTR knockout/overexpression can ameliorate the depressive-like behaviors observed in CRS-induced depressive mice. Furthermore, p75NTR knockout/overexpression safeguards the tissue morphology of the hippocampus, inhibits the mTOR signaling pathway to restore autophagy, and modulates apoptosis-related proteins (Bcl-2 and Bax) to reestablish normal levels of autophagy and apoptosis in hippocampal neurons of depressed mice. Importantly, p75NTR knockout/overexpression can improve synaptic plasticity through protecting the dendritic structure and dendritic spines of hippocampal neurons, and upregulating the expression of hippocampal synaptic-related proteins (PSD95 and SYN1). SIGNIFICANCE These findings suggest that p75NTR knockout/overexpression can alleviate CRS-induced depression-like behaviors by reinstating autophagy and suppressing apoptosis in hippocampal neurons, and enhancing hippocampal synaptic plasticity via mTOR pathway. These insights may provide potential targets for clinical treatment of depression.
Collapse
MESH Headings
- Animals
- TOR Serine-Threonine Kinases/metabolism
- Mice
- Hippocampus/metabolism
- Hippocampus/pathology
- Stress, Psychological/complications
- Stress, Psychological/metabolism
- Depression/metabolism
- Depression/etiology
- Male
- Mice, Knockout
- Signal Transduction
- Restraint, Physical
- Receptors, Nerve Growth Factor/metabolism
- Receptors, Nerve Growth Factor/genetics
- Mice, Inbred C57BL
- Behavior, Animal
- Neuronal Plasticity
- Apoptosis
- Disease Models, Animal
- Nerve Tissue Proteins/metabolism
- Nerve Tissue Proteins/genetics
- Autophagy/physiology
- Depressive Disorder, Major/metabolism
- Neurons/metabolism
- Neurons/pathology
Collapse
Affiliation(s)
- Yu-Yuan Peng
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing 400042, China; College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Can Tang
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Hai-Yan Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Yang Ding
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Huan Yang
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Xin-Mei Ma
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Jie Gao
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Sen Li
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Zai-Yun Long
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Xiu-Min Lu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China.
| | - Yong-Tang Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing 400042, China.
| |
Collapse
|
19
|
Lei M, Liu Q, Nie J, Huang R, Mei Y, Pan D, Chen Y, Liu W. Impact and Mechanisms of Action of BDNF on Neurological Disorders, Cancer, and Cardiovascular Diseases. CNS Neurosci Ther 2024; 30:e70138. [PMID: 39648800 PMCID: PMC11626086 DOI: 10.1111/cns.70138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/04/2024] [Accepted: 11/10/2024] [Indexed: 12/10/2024] Open
Abstract
Brain-derived neurotrophic factor (BDNF), which is primarily expressed in the brain and nervous tissues, is the most abundant neurotrophic factor in the adult brain. BDNF serves not only as a major neurotrophic signaling agent in the human body but also as a crucial neuromodulator. Widely distributed throughout the central nervous system (CNS), both BDNF and its receptors play a significant role in promoting neuronal survival and growth, thereby exerting neuroprotective effects. It is further considered as a guiding medium for the functionality and structural plasticity of the CNS. Increasingly, research has indicated the critical importance of BDNF in understanding human diseases. Activation of intracellular signaling pathways such as the mitogen-activated protein kinase pathway, phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin pathway, and phospholipase C γ pathway by BDNF can all potentially enhance the growth, survival, proliferation, and migration of cancer cells, influencing cancer development. The loss of BDNF and its receptor, tropomyosin receptor kinase B, in signaling pathways is also associated with increased susceptibility to brain and heart diseases. Additionally, reduced BDNF levels in both the central and peripheral systems have been closely linked to various neurogenic diseases, including neuropathic pain and psychiatric disorders. As such, this review summarizes and analyzes the impact of BDNF on neurogenic diseases, cancer, and cardiovascular diseases. This study thereby aimed to elucidate its effects on these diseases to provide new insights and approaches for their treatment.
Collapse
Affiliation(s)
- Min Lei
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical CollegeHubei University of Science and TechnologyXianningHubeiChina
| | - Qiwen Liu
- School of Basic Medical Sciences, Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical CollegeHubei University of Science and TechnologyXianningHubeiChina
| | - Jinxia Nie
- School of Basic Medical Sciences, Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical CollegeHubei University of Science and TechnologyXianningHubeiChina
| | - Rongyi Huang
- School of Basic Medical Sciences, Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical CollegeHubei University of Science and TechnologyXianningHubeiChina
| | - Yan Mei
- School of Basic Medical Sciences, Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical CollegeHubei University of Science and TechnologyXianningHubeiChina
| | - Dan Pan
- School of Basic Medical Sciences, Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical CollegeHubei University of Science and TechnologyXianningHubeiChina
| | - Yong Chen
- Xianning Central HospitalThe First Affiliated Hospital of Hubei University of Science and TechnologyXianningHubeiChina
| | - Wu Liu
- School of Basic Medical Sciences, Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical CollegeHubei University of Science and TechnologyXianningHubeiChina
| |
Collapse
|
20
|
Yang Z, Gao C, Li Z, Jiang T, Liang Y, Jiang T, Yu C, Yan S, Li P, Zhou L. The changes of tPA/PAI-1 system are associated with the ratio of BDNF/proBDNF in major depressive disorder and SSRIs antidepressant treatment. Neuroscience 2024; 559:220-228. [PMID: 39244009 DOI: 10.1016/j.neuroscience.2024.09.005] [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: 06/07/2024] [Revised: 08/20/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024]
Abstract
Increasing evidence demonstrates that brain-derived neurotrophic factor (BDNF) can be regarded as a biomarker for major depression. Our previous work found that the ratio of mature BDNF (mBDNF) to precursor-BDNF (proBDNF) was a pivotal factor in the pathogenesis of major depressive disorder (MDD). But the mechanism behind the ratio is still obscure. Tissue plasminogen activator (tPA) and plasminogen activator inhibitor-1 (PAI-1) both play essential roles in depression by regulating the ratio of BDNF/proBDNF. In present study, we analyzed BDNF, proBDNF, tPA and PAI-1 in the peripheral blood in 57 MDD patients pre- and post-treatment and in 57 healthy controls. We verified that BDNF and tPA levels were significantly decreased, whereas proBDNF and PAI-1 levels elevated obviously in MDD group pre-treatment. And after 4 weeks SSRIs treatment, the BDNF and tPA levels increased while the proBDNF and PAI-1 levels reduced. The MDD pre-treatment group had the lowest ratio of BDNF to proBDNF compared to MDD post-treatment group and control group. Though the ratio of tPA/PAI-1 in MDD pre-treatment had not reached the significance, it was still the lowest one among the three groups. The combination of tPA + PAI + BDNF showed the best diagnostic value for MDD. In summary, our data suggested that the interaction between tPA and PAI-1 implicated to the MDD and the antidepressant treatment which might through regulating the BDNF/proBDNF ratio. The combination of tPA, PAI-1 and BDNF might offer a helpful way for MDD diagnosis.
Collapse
Affiliation(s)
- Zhilan Yang
- Department of Psychiatry, The Mental Hospital of Yunnan Province, 733 Chuanjin Road, Panlong District, Kunming 650224, Yunnan, PR China
| | - Changqing Gao
- Department of Psychiatry, The Mental Hospital of Yunnan Province, 733 Chuanjin Road, Panlong District, Kunming 650224, Yunnan, PR China
| | - Zhipeng Li
- Department of Psychiatry, The Mental Hospital of Yunnan Province, 733 Chuanjin Road, Panlong District, Kunming 650224, Yunnan, PR China
| | - Tiantian Jiang
- Department of Psychiatry, The Mental Hospital of Yunnan Province, 733 Chuanjin Road, Panlong District, Kunming 650224, Yunnan, PR China
| | - Yuhang Liang
- Department of Psychiatry, The Mental Hospital of Yunnan Province, 733 Chuanjin Road, Panlong District, Kunming 650224, Yunnan, PR China
| | - Tiankai Jiang
- Department of Psychiatry, The Mental Hospital of Yunnan Province, 733 Chuanjin Road, Panlong District, Kunming 650224, Yunnan, PR China
| | - Chen Yu
- Department of Psychiatry, The Mental Hospital of Yunnan Province, 733 Chuanjin Road, Panlong District, Kunming 650224, Yunnan, PR China
| | - Shan Yan
- Institute of Biomedical Engineering, Kunming Medical University, 1168 West Chunrong Road, Chenggong District, Kunming 650500, Yunnan, PR China
| | - Peikai Li
- Department of Clinical Psychology, The Affiliated Hospital of Yunnan University, Qingnian Road, Wuhua District, Kunming 650021, Yunnan, PR China.
| | - Li Zhou
- Department of Psychiatry, The Mental Hospital of Yunnan Province, 733 Chuanjin Road, Panlong District, Kunming 650224, Yunnan, PR China.
| |
Collapse
|
21
|
Yang F, You H, Mizui T, Ishikawa Y, Takao K, Miyakawa T, Li X, Bai T, Xia K, Zhang L, Pang D, Xu Y, Zhu C, Kojima M, Lu B. Inhibiting proBDNF to mature BDNF conversion leads to ASD-like phenotypes in vivo. Mol Psychiatry 2024; 29:3462-3474. [PMID: 38762692 DOI: 10.1038/s41380-024-02595-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 04/29/2024] [Accepted: 05/03/2024] [Indexed: 05/20/2024]
Abstract
Autism Spectrum Disorders (ASD) comprise a range of early age-onset neurodevelopment disorders with genetic heterogeneity. Most ASD related genes are involved in synaptic function, which is regulated by mature brain-derived neurotrophic factor (mBDNF) and its precursor proBDNF in a diametrically opposite manner: proBDNF inhibits while mBDNF potentiates synapses. Here we generated a knock-in mouse line (BDNFmet/leu) in which the conversion of proBDNF to mBDNF is attenuated. Biochemical experiments revealed residual mBDNF but excessive proBDNF in the brain. Similar to other ASD mouse models, the BDNFmet/leu mice showed reduced dendritic arborization, altered spines, and impaired synaptic transmission and plasticity in the hippocampus. They also exhibited ASD-like phenotypes, including stereotypical behaviors and deficits in social interaction. Moreover, the plasma proBDNF/mBDNF ratio was significantly increased in ASD patients compared to normal children in a case-control study. Thus, deficits in proBDNF to mBDNF conversion in the brain may contribute to ASD-like behaviors, and plasma proBDNF/mBDNF ratio may be a potential biomarker for ASD.
Collapse
Affiliation(s)
- Feng Yang
- China National Clinical Research Center for Neurological Diseases, Basic and Translational Medicine Center, Beijing Tiantan Hospital, Capital Medical University, 100070, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, 100070, Beijing, China
| | - He You
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, 100070, Beijing, China
- School of Pharmaceutical Sciences and IDG/McGovern Institute for Brain Research, Tsinghua University, 100084, Beijing, China
| | - Toshiyuki Mizui
- Core Research for Evolutional Science and Technology (CREST), Kawaguchi, 332-0012, Japan
| | - Yasuyuki Ishikawa
- Department of Systems Life Engineering, Maebashi Institute of Technology, Maebashi, 371-0816, Japan
| | - Keizo Takao
- Core Research for Evolutional Science and Technology (CREST), Kawaguchi, 332-0012, Japan
- Life Science Research Center, University of Toyama, Toyama, 930-0194, Japan
- Department of Behavioral Physiology, Graduate School of Innovative Life Science, University of Toyama, Toyama, 930-0194, Japan
| | - Tsuyoshi Miyakawa
- Core Research for Evolutional Science and Technology (CREST), Kawaguchi, 332-0012, Japan
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Xiaofei Li
- China National Clinical Research Center for Neurological Diseases, Basic and Translational Medicine Center, Beijing Tiantan Hospital, Capital Medical University, 100070, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, 100070, Beijing, China
| | - Ting Bai
- Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Kun Xia
- Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Lingling Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Dizhou Pang
- Center for Child Behavioral Development, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yiran Xu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Masami Kojima
- Core Research for Evolutional Science and Technology (CREST), Kawaguchi, 332-0012, Japan.
- Biomedical Department of Applied Bioscience, College of Bioscience and Chemistry, Kanazawa Institute of Technology (KIT), Ishikawa, 924-0838, Japan.
| | - Bai Lu
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, 100070, Beijing, China.
- School of Pharmaceutical Sciences and IDG/McGovern Institute for Brain Research, Tsinghua University, 100084, Beijing, China.
| |
Collapse
|
22
|
Miranda M, Navas MC, Zanoni Saad MB, Piromalli Girado D, Weisstaub N, Bekinschtein P. Environmental enrichment in middle age rats improves spatial and object memory discrimination deficits. Front Behav Neurosci 2024; 18:1478656. [PMID: 39494036 PMCID: PMC11528545 DOI: 10.3389/fnbeh.2024.1478656] [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: 08/10/2024] [Accepted: 09/30/2024] [Indexed: 11/05/2024] Open
Abstract
Changes in memory performance are one of the main symptoms of normal aging. The storage of similar experiences as different memories (ie. behavioral pattern separation), becomes less efficient as aging progresses. Studies have focused on hippocampus dependent spatial memories and their role in the aging related deficits in behavioral pattern separation (BPS) by targeting high similarity interference conditions. However, parahippocampal cortices such as the perirhinal cortex are also particularly vulnerable to aging. Middle age is thought to be the stage where mild mnemonic deficits begin to emerge. Therefore, a better understanding of the timing of the spatial and object domain memory impairment could shed light over how plasticity changes in the parahipocampal-hippocampal system affects mnemonic function in early aging. In the present work, we compared the performance of young and middle-aged rats in both spatial (spontaneous location recognition) and non-spatial (spontaneous object recognition) behavioral pattern separation tasks to understand the comparative progression of these deficits from early stages of aging. Moreover, we explored the impact of environmental enrichment (EE) as an intervention with important translational value. Although a bulk of studies have examined the contribution of EE for preventing age related memory decline in diverse cognitive domains, there is limited knowledge of how this intervention could specifically impact on BPS function in middle-aged animals. Here we evaluate the effects of EE as modulator of BPS, and its ability to revert the deficits caused by normal aging at early stages. We reveal a domain-dependent impairment in behavioral pattern separation in middle-aged rats, with spatial memories affected independently of the similarity of the experiences and object memories only affected when the stimuli are similar, an effect that could be linked to the higher interference seen in this group. Moreover, we found that EE significantly enhanced behavioral performance in middle-aged rats in the spatial and object domain, and this improvement is specific of the high similarity load condition. In conclusion, these results suggest that memory is differentially affected by aging in the object and spatial domains, but that BPS function is responsive to an EE intervention in a multidomain manner.
Collapse
|
23
|
Gellé T, Vinais T, Lacroix A, Plansont B, Nubukpo P, Girard M. Serum BDNF and pro-BDNF levels in alcohol use disorders according to depression status: An exploratory study of their evolution two months after withdrawal. Heliyon 2024; 10:e38940. [PMID: 39430530 PMCID: PMC11490827 DOI: 10.1016/j.heliyon.2024.e38940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 09/25/2024] [Accepted: 10/02/2024] [Indexed: 10/22/2024] Open
Abstract
Background Alcohol use disorders (AUDs) are complex pathologies with a myriad of molecular actors involved in both disease progression and remission. Brain-derived neurotrophic factor (BDNF) is suspected to be one such actor due to its neurotrophic effects. The BDNF precursor, pro-BDNF, has different effects, as it mainly promotes neuronal apoptosis. Both forms also play a role in depression and depressive episodes (DE). The aim of this exploratory study was to compare serum BDNF and pro-BDNF levels in patients with AUDs after withdrawal and according to DE status with those of controls without AUDs or DE. Materials and methods Ninety-nine AUD patients and 40 controls were included. Questionnaires were used to assess both alcohol and psychiatric domains: the severity of hazardous alcohol consumption was assessed using Alcohol Use Disorders Identification Test (AUDIT), craving was assessed using Obsessive and Compulsive Drinking Scale (OCDS), anxiety was assessed with Hamilton Anxiety Rating Scale (HAM-A) and depression with Montgomery-Åsberg Depression Rating Scale (MADRS). Blood samples were collected during two visits: at the time of alcohol withdrawal (M0) and two months later (M2). ELISAs to measure serum BDNF and pro-BDNF levels were performed. AUD patients were categorized according to depression status at M2. Forty-five patients remained abstinent whereas 54 relapsed. BDNF serum levels rose after alcohol withdrawal, but pro-BDNF levels did not vary between M0 and M2. Results AUD subjects without DE at M2 had higher BDNF levels at both M0 and M2 than AUD subjects with DE at M2. AUD subjects showed lower MADRS and OCD scores at M2 than at M0. AUD subjects without DE had lower BDNF levels at M0 than controls but not at M2, regardless of abstinence maintenance. Conclusion BDNF serum levels were reduced in AUD patients compared to controls and were further reduced in patients with both AUDs and DE. Alcohol withdrawal treatment was sufficient to induce an increase in serum BDNF levels after 2 months, regardless of whether abstinence was maintained during this time period.
Collapse
Affiliation(s)
- Thibaut Gellé
- Inserm U1094, IRD UMR270, Univ. Limoges, CHU Limoges, EpiMaCT - Epidemiology of chronic diseases in tropical zone, Institute of Epidemiology and Tropical Neurology, OmegaHealth, Limoges, France
| | - Théodore Vinais
- Inserm U1094, IRD UMR270, Univ. Limoges, CHU Limoges, EpiMaCT - Epidemiology of chronic diseases in tropical zone, Institute of Epidemiology and Tropical Neurology, OmegaHealth, Limoges, France
- Research and Innovation Unit, Esquirol Hospital, 87025, Limoges, France
| | - Aurélie Lacroix
- Inserm U1094, IRD UMR270, Univ. Limoges, CHU Limoges, EpiMaCT - Epidemiology of chronic diseases in tropical zone, Institute of Epidemiology and Tropical Neurology, OmegaHealth, Limoges, France
- Research and Innovation Unit, Esquirol Hospital, 87025, Limoges, France
| | - Brigitte Plansont
- Research and Innovation Unit, Esquirol Hospital, 87025, Limoges, France
| | - Philippe Nubukpo
- Inserm U1094, IRD UMR270, Univ. Limoges, CHU Limoges, EpiMaCT - Epidemiology of chronic diseases in tropical zone, Institute of Epidemiology and Tropical Neurology, OmegaHealth, Limoges, France
- Research and Innovation Unit, Esquirol Hospital, 87025, Limoges, France
| | - Murielle Girard
- Research and Innovation Unit, Esquirol Hospital, 87025, Limoges, France
| |
Collapse
|
24
|
Eghbal E, Aliniaeifard S, Mehrjerdi MZ, Abdi S, Hassani SB, Rassaie T, Gruda NS. Growth, phytochemical, and phytohormonal responses of basil to different light durations and intensities under constant daily light integral. BMC PLANT BIOLOGY 2024; 24:935. [PMID: 39379825 PMCID: PMC11462769 DOI: 10.1186/s12870-024-05637-w] [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: 06/22/2024] [Accepted: 09/25/2024] [Indexed: 10/10/2024]
Abstract
Horticulture in controlled environments has been increasingly used to tackle limitations on crop production. As a crucial environmental factor, light regulate plant growth and metabolism. In the present study, basil plants were subjected to different light durations and intensities considering constant daily light integral (DLI). The lighting environment included 200, 300, and 400 µmol m- 2 s- 1 intensities for 18, 12, and 9 h, respectively. DLI amounted to 12.96 mol m- 2 d- 1 among all light treatments (LI200 for 18 h, LI300 for 12 h, and LI400 for 9 h). Half of the plants under each light treatment were exposed to 30 µmol m- 2 s- 1 of far-red light. The results indicated the general negative impact of LI400/9 on the growth of basils. Exposure to far-red light hurt the growth of the shoot, while it enhanced stem and petiole elongation. This effect was due to higher gibberellin accumulation, which resulted in shade avoidance responses. Exposure to far-red light also reduced anthocyanin and flavonoid contents, as two important nutritional components. Soluble carbohydrates increased, while storage carbohydrates decreased by increasing lighting duration/decreasing light intensity or by far-red light inclusion. The lowest antioxidant activity was detected in LI400/9. In the LI200/18, the highest level of auxin and the lowest level of cytokinin were detected, while the LI300/12 exhibited the highest level of gibberellin hormone. Low light intensity and long photoperiod enhanced plant biomass and phytochemical production and are recommended for basil production in controlled environments.
Collapse
Affiliation(s)
- Elyas Eghbal
- Photosynthesis Laboratory, Department of Horticulture, Faculty of Agricultural Technology (Aburaihan), University of Tehran, Pakdasht, Tehran, Iran
- Controlled Environment Agriculture Center (CEAC), College of Agriculture and Natural Resources, Faculty of Agricultural Technology (Aburaihan), University of Tehran, Pakdasht, Tehran, Iran
| | - Sasan Aliniaeifard
- Photosynthesis Laboratory, Department of Horticulture, Faculty of Agricultural Technology (Aburaihan), University of Tehran, Pakdasht, Tehran, Iran.
- Controlled Environment Agriculture Center (CEAC), College of Agriculture and Natural Resources, Faculty of Agricultural Technology (Aburaihan), University of Tehran, Pakdasht, Tehran, Iran.
| | - Mahboobeh Zare Mehrjerdi
- Photosynthesis Laboratory, Department of Horticulture, Faculty of Agricultural Technology (Aburaihan), University of Tehran, Pakdasht, Tehran, Iran
- Controlled Environment Agriculture Center (CEAC), College of Agriculture and Natural Resources, Faculty of Agricultural Technology (Aburaihan), University of Tehran, Pakdasht, Tehran, Iran
| | - Sahar Abdi
- Photosynthesis Laboratory, Department of Horticulture, Faculty of Agricultural Technology (Aburaihan), University of Tehran, Pakdasht, Tehran, Iran
- Controlled Environment Agriculture Center (CEAC), College of Agriculture and Natural Resources, Faculty of Agricultural Technology (Aburaihan), University of Tehran, Pakdasht, Tehran, Iran
| | - Seyedeh Batool Hassani
- Department of Plant Sciences and Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Tina Rassaie
- Department of Plant Sciences and Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Nazim S Gruda
- Department of Horticultural Science, INRES-Institute of Crop Science and Resource Conservation, University of Bonn, 53121, Bonn, Germany.
| |
Collapse
|
25
|
Garcia-Sanchez J, Lin D, Liu WW. Mechanosensitive ion channels in glaucoma pathophysiology. Vision Res 2024; 223:108473. [PMID: 39180975 PMCID: PMC11398070 DOI: 10.1016/j.visres.2024.108473] [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: 06/17/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 08/27/2024]
Abstract
Force sensing is a fundamental ability that allows cells and organisms to interact with their physical environment. The eye is constantly subjected to mechanical forces such as blinking and eye movements. Furthermore, elevated intraocular pressure (IOP) can cause mechanical strain at the optic nerve head, resulting in retinal ganglion cell death (RGC) in glaucoma. How mechanical stimuli are sensed and affect cellular physiology in the eye is unclear. Recent studies have shown that mechanosensitive ion channels are expressed in many ocular tissues relevant to glaucoma and may influence IOP regulation and RGC survival. Furthermore, variants in mechanosensitive ion channel genes may be associated with risk for primary open angle glaucoma. These findings suggest that mechanosensitive channels may be important mechanosensors mediating cellular responses to pressure signals in the eye. In this review, we focus on mechanosensitive ion channels from three major channel families-PIEZO, two-pore potassium and transient receptor potential channels. We review the key properties of these channels, their effects on cell function and physiology, and discuss their possible roles in glaucoma pathophysiology.
Collapse
Affiliation(s)
- Julian Garcia-Sanchez
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Danting Lin
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Wendy W Liu
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
26
|
Keshri PK, Singh SP. Unraveling the AKT/ERK cascade and its role in Parkinson disease. Arch Toxicol 2024; 98:3169-3190. [PMID: 39136731 DOI: 10.1007/s00204-024-03829-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/25/2024] [Indexed: 09/17/2024]
Abstract
Parkinson disease represents a significant and growing burden on global healthcare systems, necessitating a deeper understanding of their underlying molecular mechanisms for the development of effective treatments. The AKT and ERK pathways play crucial roles in the disease, influencing multiple cellular pathways that support neuronal survival. Researchers have made notable progress in uncovering how these pathways are controlled by upstream kinases and how their downstream effects contribute to cell signalling. However, as we delve deeper into their intricacies, we encounter increasing complexity, compounded by the convergence of multiple signalling pathways. Many of their targets overlap with those of other kinases, and they not only affect specific substrates but also influence entire signalling networks. This review explores the intricate interplay of the AKT/ERK pathways with several other signalling cascades, including oxidative stress, endoplasmic reticulum stress, calcium homeostasis, inflammation, and autophagy, in the context of Parkinson disease. We discuss how dysregulation of these pathways contributes to disease progression and neuronal dysfunction, highlighting potential therapeutic targets for intervention. By elucidating the complex network of interactions between the AKT/ERK pathways and other signalling cascades, this review aims to provide insights into the pathogenesis of Parkinson disease and describe the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Priyanka Kumari Keshri
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Surya Pratap Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| |
Collapse
|
27
|
Sajanti A, Li Y, Hellström S, Cao Y, Girard R, Umemori J, Frantzén J, Koskimäki F, Lyne SB, Falter J, Rantamäki T, Takala R, Posti JP, Roine S, Kolehmainen S, Srinath A, Jänkälä M, Puolitaival J, Rahi M, Rinne J, Castrén E, Koskimäki J. Brain plasticity and neuroinflammatory protein biomarkers with circulating MicroRNAs as predictors of acute brain injury outcome - A prospective cohort study. J Neurol Sci 2024; 464:123169. [PMID: 39126731 DOI: 10.1016/j.jns.2024.123169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/17/2024] [Accepted: 08/04/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND Brain recovery mechanisms after injuries like aneurysmal subarachnoid hemorrhage (aSAH), ischemic stroke (IS), and traumatic brain injury (TBI) involve brain plasticity, synaptic regeneration, and neuroinflammation. We hypothesized that serum levels of the p75 neurotrophic receptor (p75NTR) and associated signaling proteins, as well as differentially expressed (DE) microRNAs, could predict recovery outcomes irrespective of injury type. METHODS A prospective patient cohort with ischemic stroke (IS, n = 30), aneurysmal subarachnoid hemorrhage (aSAH, n = 31), and traumatic brain injury (TBI, n = 13) were evaluated (total n = 74). Serum samples were collected at two post-injury intervals (early: 1-3 days, late: 4-8 days), and outcomes were assessed after three months using the modified Rankin Scale (mRS), categorizing outcomes as favorable (mRS 0-3) or unfavorable (mRS 4-6). Six proteins were measured using ELISAs: p75NTR, NGF, sortilin, IL1β, TNFα, and cyclophilin. DE microRNAs were identified using DESeq2, and their target genes were predicted. Serum molecules between patients with differing outcomes were compared using a Kolmogorov-Smirnov test, 2-tailed t-test and multivariate linear discriminant analysis (LDA). RESULTS Favorable (n = 46) and unfavorable (n = 28) outcome cohorts were balanced with age and sex (p = 0.25 and 0.63). None of the studied proteins correlated with age. Combinatory LDA of the six protein biomarkers indicated strong prognostic value for favorable outcomes (OR 2.09; AUC = 70.3%, p = 0.0058). MicroRNA expression changes over time were identified in the aSAH, TBI, and IS groups (p < 0.05, FDR corrected). Twenty-three microRNAs were commonly DE across all brain injury groups when comparing favorable and unfavorable outcomes (p < 0.05). LDA of four microRNAs targeting the studied proteins showed high prognostic accuracy (OR 11.7; AUC = 94.1%, p = 0.016). CONCLUSIONS The combined prognostic microRNA and protein biomarker models demonstrated accurate outcome prognostication across diverse injury types, implying the presence of a common recovery mechanism. DE microRNAs were found to target the studied molecules, suggesting a potential mechanistic role in recovery. Further investigation is warranted to study these molecules in prognostication, as well as therapeutic targets for enhancing recovery.
Collapse
Affiliation(s)
- Antti Sajanti
- Neurocenter, Department of Neurosurgery, Turku University Hospital and University of Turku, P.O. Box 52, FI-20521 Turku, Finland
| | - Yan Li
- Center for Research Informatics, The University of Chicago, Chicago, IL 60637, United States of America
| | - Santtu Hellström
- Neurocenter, Department of Neurosurgery, Turku University Hospital and University of Turku, P.O. Box 52, FI-20521 Turku, Finland
| | - Ying Cao
- Department of Radiation Oncology, Kansas University Medical Center, Kansas City, KS 66160, USA
| | - Romuald Girard
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL 60637, United States of America
| | - Juzoh Umemori
- Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, FI-00014 Helsinki, Finland; Gene and Cell Technology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, 70211 Kuopio, Finland
| | - Janek Frantzén
- Neurocenter, Department of Neurosurgery, Turku University Hospital and University of Turku, P.O. Box 52, FI-20521 Turku, Finland
| | - Fredrika Koskimäki
- Neurocenter, Acute Stroke Unit, Turku University Hospital, P.O. Box 52, FI-20521 Turku, Finland
| | - Seán B Lyne
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Johannes Falter
- Department of Neurosurgery, University Medical Center of Regensburg, Regensburg 93042, Germany
| | - Tomi Rantamäki
- Laboratory of Neurotherapeutics, Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 Helsinki, Finland; SleepWell Research Program, Faculty of Medicine, University of Helsinki, P.O. Box 63, FI-00014 Helsinki, Finland
| | - Riikka Takala
- Perioperative Services, Intensive Care and Pain Medicine and Department of Anaesthesiology and Intensive Care, Turku University Hospital and University of Turku, P.O. Box52, FI-20521 Turku, Finland
| | - Jussi P Posti
- Neurocenter, Department of Neurosurgery, Turku University Hospital and University of Turku, P.O. Box 52, FI-20521 Turku, Finland
| | - Susanna Roine
- Neurocenter, Acute Stroke Unit, Turku University Hospital, P.O. Box 52, FI-20521 Turku, Finland
| | - Sulo Kolehmainen
- Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, FI-00014 Helsinki, Finland
| | - Abhinav Srinath
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL 60637, United States of America
| | - Miro Jänkälä
- Department of Neurosurgery, Oulu University Hospital, Box 25, 90029 OYS, Finland
| | - Jukka Puolitaival
- Department of Neurosurgery, Oulu University Hospital, Box 25, 90029 OYS, Finland
| | - Melissa Rahi
- Neurocenter, Department of Neurosurgery, Turku University Hospital and University of Turku, P.O. Box 52, FI-20521 Turku, Finland
| | - Jaakko Rinne
- Neurocenter, Department of Neurosurgery, Turku University Hospital and University of Turku, P.O. Box 52, FI-20521 Turku, Finland
| | - Eero Castrén
- Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, FI-00014 Helsinki, Finland
| | - Janne Koskimäki
- Neurocenter, Department of Neurosurgery, Turku University Hospital and University of Turku, P.O. Box 52, FI-20521 Turku, Finland; Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, FI-00014 Helsinki, Finland; Department of Neurosurgery, Oulu University Hospital, Box 25, 90029 OYS, Finland.
| |
Collapse
|
28
|
Staib-Lasarzik I, Gölz C, Bobkiewiecz W, Somnuke P, Sebastiani A, Thal SC, Schäfer MK. Sortilin is dispensable for secondary injury processes following traumatic brain injury in mice. Heliyon 2024; 10:e35198. [PMID: 39170542 PMCID: PMC11336488 DOI: 10.1016/j.heliyon.2024.e35198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 07/15/2024] [Accepted: 07/24/2024] [Indexed: 08/23/2024] Open
Abstract
Traumatic brain injury (TBI) is characterized by complex secondary injury processes involving the p75 neurotrophin receptor (p75NTR), which has been proposed as a possible therapeutic target. However, the pathogenic role of the p75NTR co-receptor sortilin in TBI has not been investigated. In this study, we examined whether sortilin contributes to acute and early processes of secondary injury using a murine controlled cortical impact (CCI) model of TBI. Initial expression analysis showed a down-regulation of sortilin mRNA levels 1 and 5 day post injury (dpi) and a reduced expression of sortilin protein 1 dpi. Next, a total of 40 SortilinΔExon14 loss-of-function mouse mutants (Sort1-/-) and wild-type (Sort1+/+) littermate mice were subjected to CCI and examined at 1 and 5 dpi. Neither sensorimotor deficits or brain lesion size nor CCI-induced cell death or calcium-dependent excitotoxicity as evaluated by TUNEL staining or Western blot analysis of alpha II spectrin breakdown products were different between Sort1-/- and Sort1+/+ mice. In addition, CCI induced the up-regulation of pro-inflammatory marker mRNA expression (Il6, Tnfa, Aif1, and Gfap) irrespectively of the genotype. Similarly, the mRNA expressions of neurotrophins (Bdnf, Ngf, Nt3), VPS10P domain receptors others than sortilin (Ngfr, Sorl1, Sorcs2), and the sortilin interactor progranulin were not affected by genotype. Our results suggest that sortilin is a modulatory rather than a critical factor in the acute and early brain tissue response after TBI.
Collapse
Affiliation(s)
- Irina Staib-Lasarzik
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Christina Gölz
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Wieslawa Bobkiewiecz
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Pawit Somnuke
- Department of Anesthesiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Anne Sebastiani
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Serge C. Thal
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Michael K.E. Schäfer
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Focus Program Translational Neurosciences (FTN) of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
29
|
Cao J, Gorwood P, Ramoz N, Viltart O. The Role of Central and Peripheral Brain-Derived Neurotrophic Factor (BDNF) as a Biomarker of Anorexia Nervosa Reconceptualized as a Metabo-Psychiatric Disorder. Nutrients 2024; 16:2617. [PMID: 39203753 PMCID: PMC11357464 DOI: 10.3390/nu16162617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/26/2024] [Accepted: 08/04/2024] [Indexed: 09/03/2024] Open
Abstract
Neurotrophic factors play pivotal roles in shaping brain development and function, with brain-derived neurotrophic factor (BDNF) emerging as a key regulator in various physiological processes. This review explores the intricate relationship between BDNF and anorexia nervosa (AN), a complex psychiatric disorder characterized by disordered eating behaviors and severe medical consequences. Beginning with an overview of BDNF's fundamental functions in neurodevelopment and synaptic plasticity, the review delves into recent clinical and preclinical evidence implicating BDNF in the pathophysiology of AN. Specifically, it examines the impact of BDNF polymorphisms, such as the Val66Met variant, on AN susceptibility, prognosis, and treatment response. Furthermore, the review discusses the interplay between BDNF and stress-related mood disorders, shedding light on the mechanisms underlying AN vulnerability to stress events. Additionally, it explores the involvement of BDNF in metabolic regulation, highlighting its potential implications for understanding the metabolic disturbances observed in AN. Through a comprehensive analysis of clinical data and animal studies, the review elucidates the nuanced role of BDNF in AN etiology and prognosis, emphasizing its potential as a diagnostic and prognostic biomarker. Finally, the review discusses limitations and future directions in BDNF research, underscoring the need for further investigations to elucidate the complex interplay between BDNF signaling and AN pathology.
Collapse
Affiliation(s)
- Jingxian Cao
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université Paris Cité, INSERM UMR-S 1266, F-75014 Paris, France (O.V.)
| | - Philip Gorwood
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université Paris Cité, INSERM UMR-S 1266, F-75014 Paris, France (O.V.)
- GHU Paris Psychiatrie et Neurosciences, CMME, Hôpital Sainte Anne, F-75014 Paris, France
| | - Nicolas Ramoz
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université Paris Cité, INSERM UMR-S 1266, F-75014 Paris, France (O.V.)
- GHU Paris Psychiatrie et Neurosciences, CMME, Hôpital Sainte Anne, F-75014 Paris, France
| | - Odile Viltart
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université Paris Cité, INSERM UMR-S 1266, F-75014 Paris, France (O.V.)
- SCALab Laboratory, PsySEF Faculty, Université de Lille, UMR CNRS 9193, F-59650 Villeneuve d’Ascq, France
| |
Collapse
|
30
|
Feng L, Sun J, Xia L, Shi Q, Hou Y, Zhang L, Li M, Fan C, Sun B. Ferroptosis mechanism and Alzheimer's disease. Neural Regen Res 2024; 19:1741-1750. [PMID: 38103240 PMCID: PMC10960301 DOI: 10.4103/1673-5374.389362] [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: 05/08/2023] [Revised: 07/05/2023] [Accepted: 09/09/2023] [Indexed: 12/18/2023] Open
Abstract
Regulated cell death is a genetically determined form of programmed cell death that commonly occurs during the development of living organisms. This process plays a crucial role in modulating homeostasis and is evolutionarily conserved across a diverse range of living organisms. Ferroptosis is a classic regulatory mode of cell death. Extensive studies of regulatory cell death in Alzheimer's disease have yielded increasing evidence that ferroptosis is closely related to the occurrence, development, and prognosis of Alzheimer's disease. This review summarizes the molecular mechanisms of ferroptosis and recent research advances in the role of ferroptosis in Alzheimer's disease. Our findings are expected to serve as a theoretical and experimental foundation for clinical research and targeted therapy for Alzheimer's disease.
Collapse
Affiliation(s)
- Lina Feng
- Shandong Key Laboratory of TCM Multi-Target Intervention and Disease Control, the Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Jingyi Sun
- Shandong Key Laboratory of TCM Multi-Target Intervention and Disease Control, the Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Ling Xia
- Shandong Key Laboratory of TCM Multi-Target Intervention and Disease Control, the Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Qiang Shi
- Shandong Key Laboratory of TCM Multi-Target Intervention and Disease Control, the Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Yajun Hou
- Shandong Key Laboratory of TCM Multi-Target Intervention and Disease Control, the Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Lili Zhang
- Department of Internal Medicine, Taian Traffic Hospital, Taian, Shandong Province, China
| | - Mingquan Li
- Department of Neurology, the Third Affiliated Clinical Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Cundong Fan
- Shandong Key Laboratory of TCM Multi-Target Intervention and Disease Control, the Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Baoliang Sun
- Shandong Key Laboratory of TCM Multi-Target Intervention and Disease Control, the Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| |
Collapse
|
31
|
Jang MH, Song J. Adenosine and adenosine receptors in metabolic imbalance-related neurological issues. Biomed Pharmacother 2024; 177:116996. [PMID: 38897158 PMCID: PMC12021433 DOI: 10.1016/j.biopha.2024.116996] [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: 04/24/2024] [Revised: 06/08/2024] [Accepted: 06/15/2024] [Indexed: 06/21/2024] Open
Abstract
Metabolic syndromes (e.g., obesity) are characterized by insulin resistance, chronic inflammation, impaired glucose metabolism, and dyslipidemia. Recently, patients with metabolic syndromes have experienced not only metabolic problems but also neuropathological issues, including cognitive impairment. Several studies have reported blood-brain barrier (BBB) disruption and insulin resistance in the brain of patients with obesity and diabetes. Adenosine, a purine nucleoside, is known to regulate various cellular responses (e.g., the neuroinflammatory response) by binding with adenosine receptors in the central nervous system (CNS). Adenosine has four known receptors: A1R, A2AR, A2BR, and A3R. These receptors play distinct roles in various physiological and pathological processes in the brain, including endothelial cell homeostasis, insulin sensitivity, microglial activation, lipid metabolism, immune cell infiltration, and synaptic plasticity. Here, we review the recent findings on the role of adenosine receptor-mediated signaling in neuropathological issues related to metabolic imbalance. We highlight the importance of adenosine signaling in the development of therapeutic solutions for neuropathological issues in patients with metabolic syndromes.
Collapse
Affiliation(s)
- Mi-Hyeon Jang
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, United States.
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Republic of Korea.
| |
Collapse
|
32
|
Bregman-Yemini N, Nitzan K, Franko M, Doron R. Connecting the emotional-cognitive puzzle: The role of tyrosine kinase B (TrkB) receptor isoform imbalance in age-related emotional and cognitive impairments. Ageing Res Rev 2024; 99:102349. [PMID: 38823488 DOI: 10.1016/j.arr.2024.102349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/16/2024] [Accepted: 05/26/2024] [Indexed: 06/03/2024]
Abstract
Age-related cognitive and affective disorders pose significant public health challenges. Notably, emotional and cognitive symptoms co-occur across multiple age-associated conditions like normal aging, Alzheimer's disease (AD), and mood disorders such as depression and anxiety. While the intricate interplay underlying this relationship remains poorly understood, this article highlights the possibility that an imbalance between full-length (TrkB.FL) and truncated (TrkB.T1) isoforms of tyrosine kinase receptor TrkB in the neurotrophic system may significantly affect age-associated emotional and cognitive functions, by altering brain-derived neurotrophic factor (BDNF) signaling, integral to neuronal health, cognitive functions and mood regulation. While the contribution of this imbalance to pathogenesis awaits full elucidation, this review evaluates its potential mediating role, linking emotional and cognitive decline across age-related disorders The interplay between TrkB.T1 and TrkB.FL isoforms may be considered as a pivotal shared regulator underlying this complex relationship. The current review aims to synthesize current knowledge on TrkB isoform imbalance, specifically its contribution to age-related cognitive decline and mood disorders. By examining shared pathogenic pathways between aging, cognitive decline, and mood disorders through the lens of TrkB signaling, this review uncovers potential therapeutic targets not previously considered, offering a fresh perspective on combating age-related mental health issues as well as cognitive deficits.
Collapse
Affiliation(s)
- Noa Bregman-Yemini
- Department of Education and Psychology, The Open University, Israel; Department of Psychology, The Hebrew University, Israel
| | - Keren Nitzan
- Department of Education and Psychology, The Open University, Israel
| | - Motty Franko
- Department of Education and Psychology, The Open University, Israel; Department of Psychology, Ben-Gurion University, Israel
| | - Ravid Doron
- Department of Education and Psychology, The Open University, Israel.
| |
Collapse
|
33
|
Magalhães CODE, Sousa RALD, Mendes BF, Dias IR, Pereira RRS, Pereira GC, Lee KS, Peixoto MFD, Cassilhas RC. Accumulated HIIT inhibits anxiety and depression, improves cognitive function, and memory-related proteins in the hippocampus of aged rats. Exp Brain Res 2024; 242:1871-1879. [PMID: 38864869 DOI: 10.1007/s00221-024-06869-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 05/29/2024] [Indexed: 06/13/2024]
Abstract
This study aimed to compare the effects of High-Intensity Interval Training (HIIT) performed in a single session(1xHIIT) versus three daily sessions (3xHIIT) on fitness level and behavior of aged rats. Eighteen-month-old Wistar rats were assigned to Untrained (UN), 1xHIIT, or 3xHIIT (n = 12/group). Both groups, 1xHIIT and 3xHIIT, performed 15 min of a treadmill running HIIT protocol during 8 weeks. 1xHIIT protocol consisted of a single daily session of 15 min, while the 3xHIIT performed three daily sessions of 5 min with a 4 h interval between the sessions. Morris Water Maze (MWM) task was used to evaluate spatial learning and memory. Splash test, Forced Swim test, and Elevated Plus Maze task (EPM) were used to evaluate anhedonic, depressive-like, and anxious behaviors, respectively. Rats were euthanized, and the hippocampus was harvested for western blot analyses (CaMKII and BDNF). Both HIIT protocols improved VO2max and spatial memory. Notably, only the 3xHIIT protocol attenuated anxious and depressive-like behaviors. Western blot analyses of the hippocampus revealed that both HIIT protocols increased BDNF levels. BDNF levels were higher in the 3xHIIT when compared with 1xHIIT group, and we observed increasement of the CamKII levels just in the 3x HIIT group. Therefore, this study provides evidence indicating that accumulated HIIT sessions is more effective than traditional daily HIIT sessions in improving fitness level, cognitive function, memory, inhibiting the development of mood disorders, and enhancing BDNF and CaMKII levels in the hippocampus of aged rats.
Collapse
Affiliation(s)
- Caíque Olegário Diniz E Magalhães
- Department of Physical Education, Federal University of the Jequitinhonha and Mucuri Valleys (UFVJM), Rodovia MGT 367 - Km 583, nº 5000, Diamantina, MG, CEP 39100-000, Brazil
- NNeuroscience and Exercise Study Group (Grupo de Estudos em Neurociências e Exercício - GENE), UFVJM, Diamantina, Brazil
- Multicenter Post Graduation Program in Physiological Sciences (PPGMCF), UFVJM, Brazilian Society of Physiology, Diamantina, Brazil
| | - Ricardo Augusto Leoni De Sousa
- Department of Physical Education, Federal University of the Jequitinhonha and Mucuri Valleys (UFVJM), Rodovia MGT 367 - Km 583, nº 5000, Diamantina, MG, CEP 39100-000, Brazil
- NNeuroscience and Exercise Study Group (Grupo de Estudos em Neurociências e Exercício - GENE), UFVJM, Diamantina, Brazil
- Multicenter Post Graduation Program in Physiological Sciences (PPGMCF), UFVJM, Brazilian Society of Physiology, Diamantina, Brazil
| | - Bruno Ferreira Mendes
- Department of Physical Education, Federal University of the Jequitinhonha and Mucuri Valleys (UFVJM), Rodovia MGT 367 - Km 583, nº 5000, Diamantina, MG, CEP 39100-000, Brazil
- Multicenter Post Graduation Program in Physiological Sciences (PPGMCF), UFVJM, Brazilian Society of Physiology, Diamantina, Brazil
| | - Isabela Rocha Dias
- Department of Physical Education, Federal University of the Jequitinhonha and Mucuri Valleys (UFVJM), Rodovia MGT 367 - Km 583, nº 5000, Diamantina, MG, CEP 39100-000, Brazil
- NNeuroscience and Exercise Study Group (Grupo de Estudos em Neurociências e Exercício - GENE), UFVJM, Diamantina, Brazil
- Multicenter Post Graduation Program in Physiological Sciences (PPGMCF), UFVJM, Brazilian Society of Physiology, Diamantina, Brazil
| | - Ramona Ramalho Souza Pereira
- Department of Physical Education, Federal University of the Jequitinhonha and Mucuri Valleys (UFVJM), Rodovia MGT 367 - Km 583, nº 5000, Diamantina, MG, CEP 39100-000, Brazil
- Post Graduation Program in Health Sciences (PPGCS), UFVJM, Diamantina, MG, Brazil
| | - Gabriela Cruz Pereira
- Departament of Biochemistry, Federal University of Sao Paulo (UNIFESP), São Paulo, Brazil
| | - Kil Sun Lee
- Departament of Biochemistry, Federal University of Sao Paulo (UNIFESP), São Paulo, Brazil
| | - Marco Fabricio Dias Peixoto
- Department of Physical Education, Federal University of the Jequitinhonha and Mucuri Valleys (UFVJM), Rodovia MGT 367 - Km 583, nº 5000, Diamantina, MG, CEP 39100-000, Brazil
- Multicenter Post Graduation Program in Physiological Sciences (PPGMCF), UFVJM, Brazilian Society of Physiology, Diamantina, Brazil
- Post Graduation Program in Health Sciences (PPGCS), UFVJM, Diamantina, MG, Brazil
| | - Ricardo Cardoso Cassilhas
- Department of Physical Education, Federal University of the Jequitinhonha and Mucuri Valleys (UFVJM), Rodovia MGT 367 - Km 583, nº 5000, Diamantina, MG, CEP 39100-000, Brazil.
- NNeuroscience and Exercise Study Group (Grupo de Estudos em Neurociências e Exercício - GENE), UFVJM, Diamantina, Brazil.
- Multicenter Post Graduation Program in Physiological Sciences (PPGMCF), UFVJM, Brazilian Society of Physiology, Diamantina, Brazil.
- Post Graduation Program in Health Sciences (PPGCS), UFVJM, Diamantina, MG, Brazil.
| |
Collapse
|
34
|
Wang Y, Chen S, Xue M, Ma J, Yi X, Li X, Lu X, Zhu M, Peng J, Tang Y, Zhu Y. Epigenetic regulation of key gene of PCK1 by enhancer and super-enhancer in the pathogenesis of fatty liver hemorrhagic syndrome. Anim Biosci 2024; 37:1317-1332. [PMID: 38665091 PMCID: PMC11222861 DOI: 10.5713/ab.23.0423] [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: 10/18/2023] [Revised: 01/31/2024] [Accepted: 02/18/2024] [Indexed: 07/05/2024] Open
Abstract
OBJECTIVE Rare study of the non-coding and regulatory regions of the genome limits our ability to decode the mechanisms of fatty liver hemorrhage syndrome (FLHS) in chickens. METHODS Herein, we constructed the high-fat diet-induced FLHS chicken model to investigate the genome-wide active enhancers and transcriptome by H3K27ac target chromatin immunoprecipitation sequencing (ChIP-seq) and RNA sequencing (RNA-Seq) profiles of normal and FLHS liver tissues. Concurrently, an integrative analysis combining ChIP-seq with RNA-Seq and a comparative analysis with chicken FLHS, rat non-alcoholic fatty liver disease (NAFLD) and human NAFLD at the transcriptome level revealed the enhancer and super enhancer target genes and conservative genes involved in metabolic processes. RESULTS In total, 56 and 199 peak-genes were identified in upregulated peak-genes positively regulated by H3K27ac (Cor (peak-gene correlation) ≥0.5 and log2(FoldChange) ≥1) (PP) and downregulated peak-genes positively regulated by H3K27ac (Cor (peak-gene correlation) ≥0.5 and log2(FoldChange)≤-1) (PN), respectively; then we screened key regulatory targets mainly distributing in lipid metabolism (PCK1, APOA4, APOA1, INHBE) and apoptosis (KIT, NTRK2) together with MAPK and PPAR signaling pathway in FLHS. Intriguingly, PCK1 was also significantly covered in up-regulated super-enhancers (SEs), which further implied the vital role of PCK1 during the development of FLHS. CONCLUSION Together, our studies have identified potential therapeutic biomarkers of PCK1 and elucidated novel insights into the pathogenesis of FLHS, especially for the epigenetic perspective.
Collapse
Affiliation(s)
- Yi Wang
- Department of Pathophysiology, Anhui Medical University, Hefei, 230032,
China
| | - Shuwen Chen
- Department of Pathophysiology, Anhui Medical University, Hefei, 230032,
China
| | - Min Xue
- Department of Pathophysiology, Anhui Medical University, Hefei, 230032,
China
| | - Jinhu Ma
- Department of Pathophysiology, Anhui Medical University, Hefei, 230032,
China
| | - Xinrui Yi
- Department of Pathophysiology, Anhui Medical University, Hefei, 230032,
China
| | - Xinyu Li
- Department of Pathophysiology, Anhui Medical University, Hefei, 230032,
China
| | - Xuejin Lu
- Department of Pathophysiology, Anhui Medical University, Hefei, 230032,
China
| | - Meizi Zhu
- Department of Pathophysiology, Anhui Medical University, Hefei, 230032,
China
| | - Jin Peng
- Department of Pathophysiology, Anhui Medical University, Hefei, 230032,
China
| | - Yunshu Tang
- Department of Pathophysiology, Anhui Medical University, Hefei, 230032,
China
- Laboratory Animal Research Center, College of Basic Medical Science, Anhui Medical University, Hefei, 230032,
China
| | - Yaling Zhu
- Department of Pathophysiology, Anhui Medical University, Hefei, 230032,
China
- Laboratory Animal Research Center, College of Basic Medical Science, Anhui Medical University, Hefei, 230032,
China
| |
Collapse
|
35
|
Aleksandrova E, Dimov D, Tacheva T, Petrova H, Celik K, Vlaykova T. Smoking-Dependent Association of Serum Brain-Derived Neurotrophic Factor with Pulmonary Function Parameters in Chronic Obstructive Pulmonary Disease. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1111. [PMID: 39064540 PMCID: PMC11279016 DOI: 10.3390/medicina60071111] [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: 06/05/2024] [Revised: 06/28/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024]
Abstract
Background and Objectives: One of the members of the neurotrophin (NT) family is the brain-derived neurotrophic factor (BDNF). In addition to its role in the nerve system, it has been found to play a role in lung health and diseases. Materials and Methods: The serum concentrations of BDNF were assessed in 57 patients with COPD and in 19 control individuals and the possible associations of BDNF with the spirometric indexes and disease stages were explored. Results: We did not find a significant difference between the serum concentrations of BDNF of patients and controls (p = 0.521). A significant negative correlation of the serum BDNF levels with the age of the patients (Rho = -0.279, p = 0.036) was observed. In addition, a borderline negative correlation with the age of disease onset (Rho= -0.244, p = 0.063) was also found. When analyzing these correlations in different genders, we found stronger statistical significance in male patients (Rho = -0.398, p = 0.009; and Rho = -0.419, p = 0.006), while no such significance was found in females (p = 0.574 and p = 0.342). The analyses of the possible relations of serum BDNF concentration with the spirometric parameters in the whole group of patients did not reveal any significance (p = 0.231 for FEV1%pr. and p = 0.271 for FEV1/FVC%). However, when the patients were dichotomized on the basis of smoking habits, we obtained a strong positive correlation between BDNF and FEV1%pr. (Rho = 0.501, p = 0.048) in non-smokers, but strong negative correlations with FEV1%pr. (Rho = -0.468, p = 0.003) and with FEV1/FVC% (Rho = -0.331, p = 0.040) in ex/current smokers. Non-smokers with moderate disease (GOLD II) had higher BDNF serum concentrations than patients with GOLD stage III/IV (p = 0.031). In ex/current smokers, there was an opposite association (p = 0.045). Conclusions: The results of our study suggest that the expression and secretion of BDNF are changed in COPD, but its effects and functions may differ according to the smoking history of the patients.
Collapse
Affiliation(s)
- Elina Aleksandrova
- Department of Medical Chemistry and Biochemistry, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria; (D.D.); (T.T.); (H.P.); (K.C.); (T.V.)
| | - Dimo Dimov
- Department of Medical Chemistry and Biochemistry, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria; (D.D.); (T.T.); (H.P.); (K.C.); (T.V.)
| | - Tanya Tacheva
- Department of Medical Chemistry and Biochemistry, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria; (D.D.); (T.T.); (H.P.); (K.C.); (T.V.)
| | - Hristina Petrova
- Department of Medical Chemistry and Biochemistry, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria; (D.D.); (T.T.); (H.P.); (K.C.); (T.V.)
| | - Kahan Celik
- Department of Medical Chemistry and Biochemistry, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria; (D.D.); (T.T.); (H.P.); (K.C.); (T.V.)
| | - Tatyana Vlaykova
- Department of Medical Chemistry and Biochemistry, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria; (D.D.); (T.T.); (H.P.); (K.C.); (T.V.)
- Department of Medical Biochemistry, Faculty of Pharmacy, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria
| |
Collapse
|
36
|
Pravikova PD, Arssan MA, Zalivina EA, Kondaurova EM, Kulikova EA, Belokopytova II, Naumenko VS. Dopamine receptors and key elements of the neurotrophins (BDNF, CDNF) expression patterns during critical periods of ontogenesis in the brain structures of mice with autism-like behavior (BTBR) or its absence (С57BL/6 J). Vavilovskii Zhurnal Genet Selektsii 2024; 28:407-415. [PMID: 39027124 PMCID: PMC11253014 DOI: 10.18699/vjgb-24-46] [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: 01/31/2024] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 07/20/2024] Open
Abstract
Analysis of the mechanisms underlying autism spectrum disorder (ASD) is an urgent task due to the ever-increasing prevalence of this condition. The study of critical periods of neuroontogenesis is of interest, since the manifestation of ASD is often associated with prenatal disorders of the brain development. One of the currently promising hypotheses postulates a connection between the pathogenesis of ASD and the dysfunction of neurotransmitters and neurotrophins. In this study, we investigated the expression of key dopamine receptors (Drd1, Drd2), brain-derived neurotrophic factor (Bdnf), its receptors (Ntrkb2, Ngfr) and the transcription factor Creb1 that mediates BDNF action, as well as cerebral dopamine neurotrophic factor (Cdnf) during the critical periods of embryogenesis (e14 and e18) and postnatal development (p14, p28, p60) in the hippocampus and frontal cortex of BTBR mice with autism-like behavior compared to the neurotypical C57BL/6 J strain. In BTBR embryos, on the 14th day of prenatal development, an increase in the expression of the Ngfr gene encoding the p75NTR receptor, which may lead to the activation of apoptosis, was found in the hippocampus and frontal cortex. A decrease in the expression of Cdnf, Bdnf and its receptor Ntrkb2, as well as dopamine receptors (Drd1, Drd2) was detected in BTBR mice in the postnatal period of ontogenesis mainly in the frontal cortex, while in the hippocampus of mature mice (p60), only a decrease in the Drd2 mRNA level was revealed. The obtained results suggest that the decrease in the expression levels of CDNF, BDNF-TrkB and dopamine receptors in the frontal cortex in the postnatal period can lead to significant changes in both the morphology of neurons and dopamine neurotransmission in cortical brain structures. At the same time, the increase in p75NTR receptor gene expression observed on the 14th day of embryogenesis, crucial for hippocampus and frontal cortex development, may have direct relevance to the manifestation of early autism.
Collapse
Affiliation(s)
- P D Pravikova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - M A Arssan
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - E A Zalivina
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - E M Kondaurova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - E A Kulikova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - I I Belokopytova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - V S Naumenko
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
37
|
Chang Z, Wang QY, Li LH, Jiang B, Zhou XM, Zhu H, Sun YP, Pan X, Tu XX, Wang W, Liu CY, Kuang HX. Potential Plausible Role of Stem Cell for Treating Depressive Disorder: a Retrospective Review. Mol Neurobiol 2024; 61:4454-4472. [PMID: 38097915 DOI: 10.1007/s12035-023-03843-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/29/2023] [Indexed: 07/11/2024]
Abstract
Depression poses a significant threat to global physical and mental health, impacting around 3.8% of the population with a rising incidence. Current treatment options primarily involve medication and psychological support, yet their effectiveness remains limited, contributing to high relapse rates. There is an urgent need for innovative and more efficacious treatment modalities. Stem cell therapy, a promising avenue in regenerative medicine for a spectrum of neurodegenerative conditions, has recently garnered attention for its potential application in depression. While much of this work remains preclinical, it has demonstrated considerable promise. Identified mechanisms underlying the antidepressant effects of stem cell therapy encompass the stimulation of neurotrophic factors, immune function modulation, and augmented monoamine levels. Nonetheless, these pathways and other undiscovered mechanisms necessitate further investigation. Depression fundamentally manifests as a neurodegenerative disorder. Given stem cell therapy's success in addressing a range of neurodegenerative pathologies, it opens the door to explore its application in depression treatment. This exploration may include repairing damaged nerves directly or indirectly and inhibiting neurotoxicity. Nevertheless, significant challenges must be overcome before stem cell therapies can be applied clinically. Successful resolution of these issues will ultimately determine the feasibility of incorporating stem cell therapies into the clinical landscape. This narrative review provides insights into the progress of research, potential avenues for exploration, and the prevailing challenges in the implementation of stem cell therapy for treatment of depression.
Collapse
Affiliation(s)
- Zhuo Chang
- Heilongjiang University of Chinese Medicine, Heping Road 26, Harbin, Heilongjiang, 150040, China
| | - Qing-Yi Wang
- Heilongjiang University of Chinese Medicine, Heping Road 26, Harbin, Heilongjiang, 150040, China
| | - Lu-Hao Li
- Heilongjiang University of Chinese Medicine, Heping Road 26, Harbin, Heilongjiang, 150040, China
| | - Bei Jiang
- Heilongjiang University of Chinese Medicine, Heping Road 26, Harbin, Heilongjiang, 150040, China
| | - Xue-Ming Zhou
- Heilongjiang University of Chinese Medicine, Heping Road 26, Harbin, Heilongjiang, 150040, China
| | - Hui Zhu
- Heilongjiang University of Chinese Medicine, Heping Road 26, Harbin, Heilongjiang, 150040, China
| | - Yan-Ping Sun
- Heilongjiang University of Chinese Medicine, Heping Road 26, Harbin, Heilongjiang, 150040, China
| | - Xue Pan
- Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xu-Xu Tu
- Heilongjiang University of Chinese Medicine, Heping Road 26, Harbin, Heilongjiang, 150040, China
| | - Wei Wang
- First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Chen-Yue Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hai-Xue Kuang
- Heilongjiang University of Chinese Medicine, Heping Road 26, Harbin, Heilongjiang, 150040, China.
| |
Collapse
|
38
|
Franco-Pérez J. Mechanisms Underlying Memory Impairment Induced by Fructose. Neuroscience 2024; 548:27-38. [PMID: 38679409 DOI: 10.1016/j.neuroscience.2024.04.001] [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: 01/03/2024] [Revised: 03/04/2024] [Accepted: 04/03/2024] [Indexed: 05/01/2024]
Abstract
Fructose consumption has increased over the years, especially in adolescents living in urban areas. Growing evidence indicates that daily fructose consumption leads to some pathological conditions, including memory impairment. This review summarizes relevant data describing cognitive deficits after fructose intake and analyzes the underlying neurobiological mechanisms. Preclinical experiments show sex-related deficits in spatial memory; that is, while males exhibit significant imbalances in spatial processing, females seem unaffected by dietary supplementation with fructose. Recognition memory has also been evaluated; however, only female rodents show a significant decline in the novel object recognition test performance. According to mechanistic evidence, fructose intake induces neuroinflammation, mitochondrial dysfunction, and oxidative stress in the short term. Subsequently, these mechanisms can trigger other long-term effects, such as inhibition of neurogenesis, downregulation of trophic factors and receptors, weakening of synaptic plasticity, and long-term potentiation decay. Integrating all these neurobiological mechanisms will help us understand the cellular and molecular processes that trigger the memory impairment induced by fructose.
Collapse
Affiliation(s)
- Javier Franco-Pérez
- Laboratorio Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía, Manuel Velasco Suárez, Insurgentes Sur 3877, Col. La Fama, C.P. 14269, CDMX, México, Mexico.
| |
Collapse
|
39
|
Wakasugi D, Kondo S, Ferdousi F, Mizuno S, Yada A, Tominaga K, Takahashi S, Isoda H. A rare olive compound oleacein functions as a TrkB agonist and mitigates neuroinflammation both in vitro and in vivo. Cell Commun Signal 2024; 22:309. [PMID: 38835076 PMCID: PMC11151522 DOI: 10.1186/s12964-024-01691-x] [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: 04/18/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND Neuroinflammation is widely acknowledged as a characteristic feature of almost all neurological disorders and specifically in depression- and anxiety-like disorders. In recent years, there has been significant attention on natural compounds with potent anti-inflammatory effects due to their potential in mitigating neuroinflammation and neuroplasticity. METHODS In the present study, we aimed to evaluate the neuroprotective effects of oleacein (OC), a rare secoiridoid derivative found in extra virgin olive oil. Our goal was to explore the BDNF/TrkB neurotrophic activity of OC and subsequently assess its potential for modulating neuroinflammatory response using human neuroblastoma cells (SH-SY5Y cells) and an in vivo model of depression induced by lipopolysaccharide (LPS)-mediated inflammation. RESULTS In SH-SY5Y cells, OC exhibited a significant dose-dependent increase in BDNF expression. This enhancement was absent when cells were co-treated with inhibitors of BDNF's receptor TrkB, as well as downstream molecules PI3K and MEK. Whole-transcriptomics analysis revealed that OC upregulated cell cycle-related genes under normal conditions, while downregulating inflammation-associated genes in LPS-induced conditions. Furthermore, surface plasmon resonance (SPR) assays demonstrated that OC exhibited a stronger and more stable binding affinity to TrkB compared to the positive control, 7,8-dihydroxyflavone. Importantly, bioluminescence imaging revealed that a single oral dose of OC significantly increased BDNF expression in the brains of Bdnf-IRES-AkaLuc mice. Furthermore, oral administration of OC at a dosage of 10 mg/kg body weight for 10 days significantly reduced immobility time in the tail suspension test compared to the LPS-treated group. RT-qPCR analysis revealed that OC significantly decreased the expression of pro-inflammatory cytokines Tnfα, Il6, and Il1β, while simultaneously enhancing Bdnf expression, as well as both pro and mature BDNF protein levels in mice hippocampus. These changes were comparable to those induced by the positive control antidepressant drug fluoxetine. Additionally, microarray analysis of mouse brains confirmed that OC could counteract LPS-induced inflammatory biological events. CONCLUSION Altogether, our study represents the first report on the potential antineuroinflammatory and antidepressant properties of OC via modulation of BDNF/TrkB neurotrophic activity. This finding underscores the potential of OC as a natural therapeutic agent for depression- and anxiety-related disorders.
Collapse
Affiliation(s)
- Daiki Wakasugi
- Tsukuba Life Science Innovation Program (T-LSI), University of Tsukuba, Tsukuba, Ibaraki, 305-8572, Japan
| | - Shinji Kondo
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Ibaraki, 305-8572, Japan
| | - Farhana Ferdousi
- Tsukuba Life Science Innovation Program (T-LSI), University of Tsukuba, Tsukuba, Ibaraki, 305-8572, Japan
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Ibaraki, 305-8572, Japan
- Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center (LARC) in Transborder Medical Research Center (TMRC), Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Akira Yada
- Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, 305-0821, Japan
- Interdisciplinary Research Center for Catalytic Chemistry, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba Central 5, 1-1-1 Higashi, Ibaraki, 305-8565, Japan
| | - Kenichi Tominaga
- Tsukuba Life Science Innovation Program (T-LSI), University of Tsukuba, Tsukuba, Ibaraki, 305-8572, Japan
- Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, 305-0821, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center (LARC) in Transborder Medical Research Center (TMRC), Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Hiroko Isoda
- Tsukuba Life Science Innovation Program (T-LSI), University of Tsukuba, Tsukuba, Ibaraki, 305-8572, Japan.
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Ibaraki, 305-8572, Japan.
- Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan.
- Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, 305-0821, Japan.
| |
Collapse
|
40
|
Kipp BT, Savage LM. Modulation of the p75NTR during Adolescent Alcohol Exposure Prevents Cholinergic Neuronal Atrophy and Associated Acetylcholine Activity and Behavioral Dysfunction. Int J Mol Sci 2024; 25:5792. [PMID: 38891978 PMCID: PMC11172149 DOI: 10.3390/ijms25115792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Binge alcohol consumption during adolescence can produce lasting deficits in learning and memory while also increasing the susceptibility to substance use disorders. The adolescent intermittent ethanol (AIE) rodent model mimics human adolescent binge drinking and has identified the nucleus basalis magnocellularis (NbM) as a key site of pathology. The NbM is a critical regulator of prefrontal cortical (PFC) cholinergic function and attention. The cholinergic phenotype is controlled pro/mature neurotrophin receptor activation. We sought to determine if p75NTR activity contributes to the loss of cholinergic phenotype in AIE by using a p75NTR modulator (LM11A-31) to inhibit prodegenerative signaling during ethanol exposure. Male and female rats underwent 5 g/kg ethanol (AIE) or water (CON) exposure following 2-day-on 2-day-off cycles from postnatal day 25-57. A subset of these groups also received a protective dose of LM11A-31 (50 mg/kg) during adolescence. Rats were trained on a sustained attention task (SAT) and behaviorally relevant acetylcholine (ACh) activity was recorded in the PFC with a fluorescent indicator (AChGRAB 3.0). AIE produced learning deficits on the SAT, which were spared with LM11A-31. In addition, PFC ACh activity was blunted by AIE, which LM11A-31 corrected. Investigation of NbM ChAT+ and TrkA+ neuronal expression found that AIE led to a reduction of ChAT+TrkA+ neurons, which again LM11A-31 protected. Taken together, these findings demonstrate the p75NTR activity during AIE treatment is a key regulator of cholinergic degeneration.
Collapse
Affiliation(s)
| | - Lisa M. Savage
- Department of Psychology, Binghamton University-State University of New York, Binghamton, NY 13902, USA;
| |
Collapse
|
41
|
Yang XP, Dan-Dai, Chen RX, Li YX, Rui Lv X, Li Y. The expression of ProBDNF and its high affinity receptor P75NTR in the neurons of emotion-related brain regions of post-stroke depression rats. Brain Res 2024; 1831:148829. [PMID: 38423239 DOI: 10.1016/j.brainres.2024.148829] [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: 12/21/2023] [Revised: 02/13/2024] [Accepted: 02/23/2024] [Indexed: 03/02/2024]
Abstract
OBJECTIVE To investigate the expression of the precursor of brain-derived neurotrophic factor (proBDNF) and its high-affinity receptor p75NTR in neurons of emotion-related brain areas (prefrontal cortex, hippocampus, and amygdala) in rats with post-stroke depression (PSD), and to explore the expression levels of proBDNF and p75NTR in neurons of emotion-related brain areas by injecting tissue plasminogen activator (t-PA) into the lateral ventricle of PSD rats, this significantly improved the stress-induced depression-like behavior,thus further validating the above results. METHODS Rats were randomly divided into four groups: a normal control group (n = 8), a depression group (n = 8), a stroke group (n = 8), and a PSD group (n = 8). The rat model of stroke was established by thread embolism, and the PSD animal model was induced by chronic unpredictable mild stress (CUMS) and solitary feeding. Behavioral tests were conducted, including weight measurement, open field tests, and sucrose preference tests. Immunofluorescence double labeling was used to detect the expression of proBDNF and p75NTR in neurons of emotion-related brain regions in the PSD rat model. Four weeks after CUMS treatment, the PSD group was selected. Rats were infused with t-PA (3 μg dissolved in 6 μL saline, Boehringer Ingelheim), proBDNF (3 μg dissolved in 6 μL saline, Abcam), or equal-volume NS once per day for 7 consecutive days using the syringe pump connecting to injection needles. After 7 days of continuous administration, animal behavior was assessed through scoring, and the expression of proBDNF and p75NTR in the emotion-related brain regions of the PSD rat model was detected using immunofluorescence double labeling. RESULTS Compared with the normal control group and the stroke group, the body weight, sucrose water consumption, and vertical movement distance in the PSD group were significantly lower (P < 0.05). In contrast, when compared with the proBDNF injection group and saline injection group, the weight, sucrose water consumption, field horizontal movement, and vertical movement distance of the t-PA injection group significantly increased after PSD lateral ventricle intubation.Double immunofluorescence revealed a higher neuronal expression of proBDNF as well as p75NTR in the prefrontal cortex and hippocampus of PSD rats compared to control animals (P < 0.05). In the amygdala, the expression levels of proBDNF and P75NTR were significantly reduced in the PSD group compared to the control group (P < 0.05). The results of the expression levels of proBDNF and P75NTR in the emotion-related brain regions of PSD rats injected with t-PA showed that proBDNF and P75NTR was significantly reduced in the prefrontal cortex, hippocampus, and amygdala of PSD rats compared to those of the NS and proBDNF groups (P < 0.05). CONCLUSIONS The increased expression of the brain-derived neurotrophic factor precursor proBDNF and its receptor p75NTR in neurons of emotion-related brain regions may play an important role in the pathogenesis of PSD.t-PA reduced the expression of proBDNF and its receptor p75NTR in neurons emotion-related brain regions and significantly improved the stress-induced depression-like behavior. Therefore, it is reasonable to assume that exogenous injection of t-PA may alleviate the depressive symptoms of PSD patients.Reducing the expression of proBDNF by injecting t-PA may provide a novel therapeutic approach for the treatment of stress-related mood disorders.
Collapse
Affiliation(s)
- Xue-Ping Yang
- Clinical Medical School, Dali University, Dali, China
| | - Dan-Dai
- Changshu Hospital Affiliated to Soochow University, Changshu No.1 People's Hospital, Changshu 215500, Jiangsu Province, China
| | - Ruo-Xia Chen
- Clinical Medical School, Dali University, Dali, China
| | - Yu-Xuan Li
- Clinical Medical School, Dali University, Dali, China
| | - Xue Rui Lv
- Clinical Medical School, Dali University, Dali, China
| | - Yun Li
- Clinical Medical School, Dali University, Dali, China; Changshu Hospital Affiliated to Soochow University, Changshu No.1 People's Hospital, Changshu 215500, Jiangsu Province, China.
| |
Collapse
|
42
|
Lee HW, Chen SJ, Tsai KJ, Hsu KS, Chen YF, Chang CH, Lin HH, Hsueh WY, Hsieh HP, Lee YF, Chiang HC, Chang JY. Targeting cathepsin S promotes activation of OLF1-BDNF/TrkB axis to enhance cognitive function. J Biomed Sci 2024; 31:46. [PMID: 38725007 PMCID: PMC11084077 DOI: 10.1186/s12929-024-01037-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 04/27/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Cathepsin S (CTSS) is a cysteine protease that played diverse roles in immunity, tumor metastasis, aging and other pathological alterations. At the cellular level, increased CTSS levels have been associated with the secretion of pro-inflammatory cytokines and disrupted the homeostasis of Ca2+ flux. Once CTSS was suppressed, elevated levels of anti-inflammatory cytokines and changes of Ca2+ influx were observed. These findings have inspired us to explore the potential role of CTSS on cognitive functions. METHODS We conducted classic Y-maze and Barnes Maze tests to assess the spatial and working memory of Ctss-/- mice, Ctss+/+ mice and Ctss+/+ mice injected with the CTSS inhibitor (RJW-58). Ex vivo analyses including long-term potentiation (LTP), Golgi staining, immunofluorescence staining of sectioned whole brain tissues obtained from experimental animals were conducted. Furthermore, molecular studies were carried out using cultured HT-22 cell line and primary cortical neurons that treated with RJW-58 to comprehensively assess the gene and protein expressions. RESULTS Our findings reported that targeting cathepsin S (CTSS) yields improvements in cognitive function, enhancing both working and spatial memory in behavior models. Ex vivo studies showed elevated levels of long-term potentiation levels and increased synaptic complexity. Microarray analysis demonstrated that brain-derived neurotrophic factor (BDNF) was upregulated when CTSS was knocked down by using siRNA. Moreover, the pharmacological blockade of the CTSS enzymatic activity promoted BDNF expression in a dose- and time-dependent manner. Notably, the inhibition of CTSS was associated with increased neurogenesis in the murine dentate gyrus. These results suggested a promising role of CTSS modulation in cognitive enhancement and neurogenesis. CONCLUSION Our findings suggest a critical role of CTSS in the regulation of cognitive function by modulating the Ca2+ influx, leading to enhanced activation of the BDNF/TrkB axis. Our study may provide a novel strategy for improving cognitive function by targeting CTSS.
Collapse
Affiliation(s)
- Hao-Wei Lee
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
- Taipei Cancer Center, TMU Research Center of Cancer Translational Medicine, Taipei Medical University Hospital, College of Medicine, Taipei Medical University, No. 252, Wuxing St., Xinyi Dist., Taipei, 110301, Taiwan (R.O.C.)
| | - Szu-Jung Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Kuen-Jer Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Research Center of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kuei-Sen Hsu
- Institute of Basic Medical Science, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Fan Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Chih-Hua Chang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Hsiao-Han Lin
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Wen-Yun Hsueh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Hsing-Pang Hsieh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Yueh-Feng Lee
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Huai-Chueh Chiang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Jang-Yang Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan.
- Taipei Cancer Center, TMU Research Center of Cancer Translational Medicine, Taipei Medical University Hospital, College of Medicine, Taipei Medical University, No. 252, Wuxing St., Xinyi Dist., Taipei, 110301, Taiwan (R.O.C.).
| |
Collapse
|
43
|
Ali NH, Al‐Kuraishy HM, Al‐Gareeb AI, Alexiou A, Papadakis M, AlAseeri AA, Alruwaili M, Saad HM, Batiha GE. BDNF/TrkB activators in Parkinson's disease: A new therapeutic strategy. J Cell Mol Med 2024; 28:e18368. [PMID: 38752280 PMCID: PMC11096816 DOI: 10.1111/jcmm.18368] [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: 10/18/2023] [Revised: 03/22/2024] [Accepted: 04/17/2024] [Indexed: 05/18/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder of the brain and is manifested by motor and non-motor symptoms because of degenerative changes in dopaminergic neurons of the substantia nigra. PD neuropathology is associated with mitochondrial dysfunction, oxidative damage and apoptosis. Thus, the modulation of mitochondrial dysfunction, oxidative damage and apoptosis by growth factors could be a novel boulevard in the management of PD. Brain-derived neurotrophic factor (BDNF) and its receptor tropomyosin receptor kinase type B (TrkB) are chiefly involved in PD neuropathology. BDNF promotes the survival of dopaminergic neurons in the substantia nigra and enhances the functional activity of striatal neurons. Deficiency of the TrkB receptor triggers degeneration of dopaminergic neurons and accumulation of α-Syn in the substantia nigra. As well, BDNF/TrkB signalling is reduced in the early phase of PD neuropathology. Targeting of BDNF/TrkB signalling by specific activators may attenuate PD neuropathology. Thus, this review aimed to discuss the potential role of BDNF/TrkB activators against PD. In conclusion, BDNF/TrkB signalling is decreased in PD and linked with disease severity and long-term complications. Activation of BDNF/TrkB by specific activators may attenuate PD neuropathology.
Collapse
Affiliation(s)
- Naif H. Ali
- Department of Internal Medicine, Medical CollegeNajran UniversityNajranSaudi Arabia
| | - Hayder M. Al‐Kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
| | | | - Athanasios Alexiou
- University Centre for Research and Development, Chandigarh UniversityMohaliPunjabIndia
- Department of Research and DevelopmentFunogenAthensGreece
- Department of Research and DevelopmentAFNP MedWienAustria
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamNew South WalesAustralia
| | - Marios Papadakis
- Department of Surgery IIUniversity Hospital Witten‐Herdecke, University of Witten‐HerdeckeWuppertalGermany
| | - Ali Abdullah AlAseeri
- Department of Internal MedicineCollege of Medicine, Prince Sattam bin Abdulaziz UniversityAl‐KharjSaudi Arabia
| | - Mubarak Alruwaili
- Department of Internal Medicine, College of MedicineJouf UniversitySakakaSaudi Arabia
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary MedicineMatrouh UniversityMatrouhEgypt
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityDamanhourEgypt
| |
Collapse
|
44
|
Shahi S, Kang T, Fonseka P. Extracellular Vesicles in Pathophysiology: A Prudent Target That Requires Careful Consideration. Cells 2024; 13:754. [PMID: 38727289 PMCID: PMC11083420 DOI: 10.3390/cells13090754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Extracellular vesicles (EVs) are membrane-bound particles released by cells to perform multitudes of biological functions. Owing to their significant implications in diseases, the pathophysiological role of EVs continues to be extensively studied, leading research to neglect the need to explore their role in normal physiology. Despite this, many identified physiological functions of EVs, including, but not limited to, tissue repair, early development and aging, are attributed to their modulatory role in various signaling pathways via intercellular communication. EVs are widely perceived as a potential therapeutic strategy for better prognosis, primarily through utilization as a mode of delivery vehicle. Moreover, disease-associated EVs serve as candidates for the targeted inhibition by pharmacological or genetic means. However, these attempts are often accompanied by major challenges, such as off-target effects, which may result in adverse phenotypes. This renders the clinical efficacy of EVs elusive, indicating that further understanding of the specific role of EVs in physiology may enhance their utility. This review highlights the essential role of EVs in maintaining cellular homeostasis under different physiological settings, and also discusses the various aspects that may potentially hinder the robust utility of EV-based therapeutics.
Collapse
Affiliation(s)
| | | | - Pamali Fonseka
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia; (S.S.); (T.K.)
| |
Collapse
|
45
|
Koning R, van Roon MA, Brouwer MC, van de Beek D. Adjunctive treatments for pneumococcal meningitis: a systematic review of experimental animal models. Brain Commun 2024; 6:fcae131. [PMID: 38707710 PMCID: PMC11069119 DOI: 10.1093/braincomms/fcae131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/22/2023] [Accepted: 04/10/2024] [Indexed: 05/07/2024] Open
Abstract
New treatments are needed to improve the prognosis of pneumococcal meningitis. We performed a systematic review on adjunctive treatments in animal models of pneumococcal meningitis in order to identify treatments with the most potential to progress to clinical trials. Studies testing therapy adjunctive to antibiotics in animal models of pneumococcal meningitis were included. A literature search was performed using Medline, Embase and Scopus for studies published from 1990 up to 17 February 2023. Two investigators screened studies for inclusion and independently extracted data. Treatment effect was assessed on the clinical parameters disease severity, hearing loss and cognitive impairment and the biological parameters inflammation, brain injury and bacterial load. Adjunctive treatments were evaluated by their effect on these outcomes and the quality, number and size of studies that investigated the treatments. Risk of bias was assessed with the SYRCLE risk of bias tool. A total of 58 of 2462 identified studies were included, which used 2703 experimental animals. Disease modelling was performed in rats (29 studies), rabbits (13 studies), mice (12 studies), gerbils (3 studies) or both rats and mice (1 study). Meningitis was induced by injection of Streptococcus pneumoniae into the subarachnoid space. Randomization of experimental groups was performed in 37 of 58 studies (64%) and 12 studies (12%) were investigator-blinded. Overall, 54 treatment regimens using 46 adjunctive drugs were evaluated: most commonly dexamethasone (16 studies), daptomycin (5 studies), complement component 5 (C5; 3 studies) antibody and Mn(III)tetrakis(4-benzoicacid)porphyrin chloride (MnTBAP; 3 studies). The most frequently evaluated outcome parameters were inflammation [32 studies (55%)] and brain injury [32 studies (55%)], followed by disease severity [30 studies (52%)], hearing loss [24 studies (41%)], bacterial load [18 studies (31%)] and cognitive impairment [9 studies (16%)]. Adjunctive therapy that improved clinical outcomes in multiple studies was dexamethasone (6 studies), C5 antibodies (3 studies) and daptomycin (3 studies). HMGB1 inhibitors, matrix metalloproteinase inhibitors, neurotrophins, antioxidants and paquinimod also improved clinical parameters but only in single or small studies. Evaluating the treatment effect of adjunctive therapy was complicated by study heterogeneity regarding the animal models used and outcomes reported. In conclusion, 24 of 54 treatment regimens (44%) tested improved clinically relevant outcomes in experimental pneumococcal meningitis but few were tested in multiple well-designed studies. The most promising new adjunctive treatments are with C5 antibodies or daptomycin, suggesting that these drugs could be tested in clinical trials.
Collapse
Affiliation(s)
- Rutger Koning
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, 1100DD Amsterdam, The Netherlands
| | - Marian A van Roon
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, 1100DD Amsterdam, The Netherlands
| | - Matthijs C Brouwer
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, 1100DD Amsterdam, The Netherlands
| | - Diederik van de Beek
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, 1100DD Amsterdam, The Netherlands
| |
Collapse
|
46
|
Zhivkovich M, Ermolaeva EV, Soboleva AV, Samoilova EM, Chudakova DA, Baklaushev VP. Brain neurotrophic factor BDNF: new data, functions and questions. GENES & CELLS 2024; 19:61-84. [DOI: 10.17816/gc623163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2025]
Abstract
The brain-derived neurotrophic factor (BDNF) is a key modulator of neurogenesis, synaptogenesis, neuroregeneration, and cell differentiation in the nervous system. Impaired BDNF functioning is a characteristic of various neurological diseases, such as Alzheimer’s disease, multiple sclerosis, and depressive disorders. There is recent evidence that patients with COVID-19 have reduced BDNF levels in the blood plasma. Furthermore, exogenous BDNF and its mimetics have demonstrated therapeutic potential.
In this review, we systematized data of the BDNF gene structure, epigenetic and microRNA-mediated regulation of its expression, transcriptional variants of BDNF, and the effects of BDNF on neuronal and oligodendroglial differentiation. Further, we point out the gaps in the current knowledge about BDNF and propose experiments that can expand such knowledge and the range of possibilities for using BDNF in biomedicine. These include determining the expression pattern of all BDNF gene transcripts at different stages of differentiation and in different cell subpopulations and studying the role of receptor-independent BDNF signaling, circadian fluctuations in BDNF levels, and their role in physiological and pathophysiological conditions. Finally, for translational medicine, evaluating the effect of BDNF mimetics (including those immobilized on three-dimensional scaffolds for tissue engineering) on neuronal and oligodendroglial differentiation of pluripotent and polypotent cells and identifying molecular regulators of BDNF transcription, including small molecules and microRNAs capable of regulating BDNF gene expression, are crucial.
Collapse
|
47
|
Speidell A, Agbey C, Mocchetti I. Accelerated neurodegeneration of basal forebrain cholinergic neurons in HIV-1 gp120 transgenic mice: Critical role of the p75 neurotrophin receptor. Brain Behav Immun 2024; 117:347-355. [PMID: 38266662 PMCID: PMC10935610 DOI: 10.1016/j.bbi.2024.01.215] [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: 09/08/2023] [Revised: 01/19/2024] [Accepted: 01/20/2024] [Indexed: 01/26/2024] Open
Abstract
Human Immunodeficiency Virus-1 (HIV) infection of the brain induces HIV-associated neurocognitive disorders (HAND). The set of molecular events employed by HIV to drive cognitive impairments in people living with HIV are diverse and remain not completely understood. We have shown that the HIV envelope protein gp120 promotes loss of synapses and decreases performance on cognitive tasks through the p75 neurotrophin receptor (p75NTR). This receptor is abundant on cholinergic neurons of the basal forebrain and contributes to cognitive impairment in various neurological disorders. In this study, we examined cholinergic neurons of gp120 transgenic (gp120tg) mice for signs of degeneration. We observed that the number of choline acetyltransferase-expressing cells is decreased in old (12-14-month-old) gp120tg mice when compared to age matched wild type. In the same animals, we observed an increase in the levels of pro-nerve growth factor, a ligand of p75NTR, as well as a disruption of consolidation of extinction of conditioned fear, a behavior regulated by cholinergic neurons of the basal forebrain. Both biochemical and behavioral outcomes of gp120tg mice were rescued by the deletion of the p75NTR gene, strongly supporting the role that this receptor plays in the neurotoxic effects of gp120. These data indicate that future p75NTR-directed pharmacotherapies could provide an adjunct therapy against synaptic simplification caused by HIV.
Collapse
Affiliation(s)
- Andrew Speidell
- Interdisciplinary Program in Neuroscience, and Department of Neuroscience, NRB WP13, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Christy Agbey
- Interdisciplinary Program in Neuroscience, and Department of Neuroscience, NRB WP13, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Italo Mocchetti
- Interdisciplinary Program in Neuroscience, and Department of Neuroscience, NRB WP13, Georgetown University Medical Center, Washington, DC 20057, USA.
| |
Collapse
|
48
|
Theik NWY, Muminovic M, Alvarez-Pinzon AM, Shoreibah A, Hussein AM, Raez LE. NTRK Therapy among Different Types of Cancers, Review and Future Perspectives. Int J Mol Sci 2024; 25:2366. [PMID: 38397049 PMCID: PMC10889397 DOI: 10.3390/ijms25042366] [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: 01/03/2024] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Neurotrophic tyrosine receptor kinase (NTRK) has been a remarkable therapeutic target for treating different malignancies, playing an essential role in oncogenic signaling pathways. Groundbreaking trials like NAVIGATE led to the approval of NTRK inhibitors by the Food and Drug Administration (FDA) to treat different malignancies, significantly impacting current oncology treatment. Accurate detection of NTRK gene fusion becomes very important for possible targeted therapy. Various methods to detect NTRK gene fusion have been applied widely based on sensitivity, specificity, and accessibility. The utility of different tests in clinical practice is discussed in this study by providing insights into their effectiveness in targeting patients who may benefit from therapy. Widespread use of NTRK inhibitors in different malignancies could remain limited due to resistance mechanisms that cause challenges to medication efficacy in addition to common side effects of the medications. This review provides a succinct overview of the application of NTRK inhibitors in various types of cancer by emphasizing the critical clinical significance of NTRK fusion gene detection. The discussion also provides a solid foundation for understanding the current challenges and potential changes for improving the efficacy of NTRK inhibitor therapy to treat different malignancies.
Collapse
Affiliation(s)
- Nyein Wint Yee Theik
- Division of Internal Medicine, Memorial Healthcare System, Pembroke Pines, FL 33028, USA; (N.W.Y.T.); (A.S.)
| | - Meri Muminovic
- Memorial Cancer Institute, Memorial Healthcare System, Pembroke Pines, FL 33028, USA;
| | - Andres M. Alvarez-Pinzon
- Memorial Cancer Institute, Office of Human Research, Florida Atlantic University (FAU), Pembroke Pines, FL 33028, USA
| | - Ahmed Shoreibah
- Division of Internal Medicine, Memorial Healthcare System, Pembroke Pines, FL 33028, USA; (N.W.Y.T.); (A.S.)
| | - Atif M. Hussein
- Memorial Cancer Institute, Memorial Healthcare System, Florida Atlantic University (FAU), Pembroke Pines, FL 33028, USA;
| | - Luis E. Raez
- Memorial Cancer Institute, Memorial Healthcare System, Florida Atlantic University (FAU), Pembroke Pines, FL 33028, USA;
| |
Collapse
|
49
|
Kim J, Seol S, Kim TE, Lee J, Koo JW, Kang HJ. Synaptotagmin-4 induces anhedonic responses to chronic stress via BDNF signaling in the medial prefrontal cortex. Exp Mol Med 2024; 56:329-343. [PMID: 38297157 PMCID: PMC10907712 DOI: 10.1038/s12276-024-01156-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 11/22/2023] [Accepted: 12/08/2023] [Indexed: 02/02/2024] Open
Abstract
Stressful circumstances are significant contributors to mental illnesses such as major depressive disorder. Anhedonia, defined as loss of the ability to enjoy pleasure in pleasurable situations, including rewarding activities or social contexts, is considered a key symptom of depression. Although stress-induced depression is associated with anhedonia in humans and animals, the underlying molecular mechanisms of anhedonic responses remain poorly understood. In this study, we demonstrated that synaptotagmin-4 (SYT4), which is involved in the release of neurotransmitters and neurotrophic factors, is implicated in chronic stress-induced anhedonia. Employing chronic unpredictable stress (CUS), we evaluated two subpopulations of mice, susceptible (SUS, anhedonic) and resilient (RES, nonanhedonic), based on sucrose preference, which was strongly correlated with social reward. The FosTRAP (targeted recombination in active populations) system and optogenetic approach revealed that neural activity in the medial prefrontal cortex (mPFC) was significantly associated with CUS-induced anhedonic behavioral phenotypes. By conducting weighted gene coexpression network analysis of RNA sequencing data from the mPFC of SUS and RES mice, we identified Syt4 as a hub gene in a gene network that was unique to anhedonia. We also confirmed that Syt4 overexpression in the mPFC was pro-susceptible, while Syt4 knockdown was pro-resilient; the pro-susceptible effects of SYT4 were mediated through a reduction in brain-derived neurotrophic factor (BDNF)-tropomyosin receptor kinase B (TrkB) signaling in the mPFC. These findings suggest that SYT4-BDNF interactions in the mPFC represent a crucial regulatory mechanism of anhedonic susceptibility to chronic stress.
Collapse
Affiliation(s)
- Jeongseop Kim
- Emotion, Cognition & Behavior Research Group, Korea Brain Research Institute (KBRI), Dong-gu, Daegu, 41062, Republic of Korea
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Dalseong-gun, Daegu, 42988, Republic of Korea
| | - Sihwan Seol
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Tae-Eun Kim
- Emotion, Cognition & Behavior Research Group, Korea Brain Research Institute (KBRI), Dong-gu, Daegu, 41062, Republic of Korea
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Dalseong-gun, Daegu, 42988, Republic of Korea
| | - Joonhee Lee
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Ja Wook Koo
- Emotion, Cognition & Behavior Research Group, Korea Brain Research Institute (KBRI), Dong-gu, Daegu, 41062, Republic of Korea.
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Dalseong-gun, Daegu, 42988, Republic of Korea.
| | - Hyo Jung Kang
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|
50
|
Xiang S, Lu X. Selective type II TRK inhibitors overcome xDFG mutation mediated acquired resistance to the second-generation inhibitors selitrectinib and repotrectinib. Acta Pharm Sin B 2024; 14:517-532. [PMID: 38322338 PMCID: PMC10840435 DOI: 10.1016/j.apsb.2023.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/15/2023] [Accepted: 10/24/2023] [Indexed: 02/08/2024] Open
Abstract
Neurotrophic receptor kinase (NTRK) fusions are actionable oncogenic drivers of multiple pediatric and adult solid tumors, and tropomyosin receptor kinase (TRK) has been considered as an attractive therapeutic target for "pan-cancer" harboring these fusions. Currently, two generations TRK inhibitors have been developed. The representative second-generation inhibitors selitrectinib and repotrectinib were designed to overcome clinic acquired resistance of the first-generation inhibitors larotrectinib or entrectinib resulted from solvent-front and gatekeeper on-target mutations. However, xDFG (TRKAG667C/A/S, homologous TRKCG696C/A/S) and some double mutations still confer resistance to selitrectinib and repotrectinib, and overcoming these resistances represents a major unmet clinical need. In this review, we summarize the acquired resistance mechanism of the first- and second-generation TRK inhibitors, and firstly put forward the emerging selective type II TRK inhibitors to overcome xDFG mutations mediated resistance. Additionally, we concluded our perspectives on new challenges and future directions in this field.
Collapse
Affiliation(s)
- Shuang Xiang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Xiaoyun Lu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| |
Collapse
|