1
|
Gupta AK, Gupta S, Mehan S, Khan Z, Das Gupta G, Narula AS. Exploring the Connection Between BDNF/TrkB and AC/cAMP/PKA/CREB Signaling Pathways: Potential for Neuroprotection and Therapeutic Targets for Neurological Disorders. Mol Neurobiol 2025:10.1007/s12035-025-05001-5. [PMID: 40342191 DOI: 10.1007/s12035-025-05001-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 04/24/2025] [Indexed: 05/11/2025]
Abstract
The BDNF/TrkB and AC/cAMP/PKA/CREB signaling pathways play a vital role in neuroplasticity, neuronal survival, and cognitive functions. This review explores its physiological and pathological implications in neurological disorders, with a focus on neurodegenerative diseases (NDDs) and neuropsychiatric disorders (NPDs). Neurological conditions increasingly burden public health, making understanding the biochemical mechanisms that underpin these diseases critical. BDNF, a neurotrophic factor, binds to the TrkB receptor, activating multiple intracellular signaling cascades that regulate cellular responses essential for neurogenesis, memory, and learning. Dysregulation within this pathway has been linked to various NDDs, as well as NPDs. Key components of the path, including adenylyl cyclase and cyclic AMP, mediate the effects of neurotransmitters and growth factors, influencing downstream targets like PKA and CREB, which are crucial for gene expression and synaptic changes. Furthermore, the review discusses the challenges of targeting this pathway for therapeutic interventions, including receptor isoform diversity, blood-brain barrier penetration, and potential side effects. Future strategies may include the development of selective TrkB modulators, nanoparticle carriers for drug delivery, and innovative gene therapy techniques. Advancing the understanding of this complex signaling network holds promise for effective interventions in treating neurological and psychiatric disorders, ultimately enhancing neuroprotection and cognitive resilience.
Collapse
Affiliation(s)
- Abhishek Kumar Gupta
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
- Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India
| | - Sumedha Gupta
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
- Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India.
- Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India.
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
- Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India
| | - Ghanshyam Das Gupta
- Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC, 27516, USA
| |
Collapse
|
2
|
Badia-Soteras A, Mak A, Blok TM, Boers-Escuder C, van den Oever MC, Min R, Smit AB, Verheijen MHG. Astrocyte-synapse structural plasticity in neurodegenerative and neuropsychiatric diseases. Biol Psychiatry 2025:S0006-3223(25)01125-4. [PMID: 40254258 DOI: 10.1016/j.biopsych.2025.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 03/18/2025] [Accepted: 04/11/2025] [Indexed: 04/22/2025]
Abstract
Synaptic dysfunction is a common feature across a broad spectrum of brain diseases, spanning from psychopathologies such as post-traumatic stress disorder (PTSD) and substance use disorders (SUD) to neurodegenerative diseases like Alzheimer's and Parkinson's disease (AD and PD). While neuroscience research aiming to understand the mechanisms underlying synaptic dysfunction has traditionally focused on the neuronal elements of the synapse, recent research increasingly acknowledges the contribution of astrocytes as a third element controlling synaptic transmission. This also sparked interest to investigate the tripartite synapse and its role in the etiology of neurological diseases. According to recent evidence, changes in the structural interaction between astrocytes and synapses not only play a pivotal role in modulating synaptic function and behavioral states, but are also implicated in the initiation and progression of various brain diseases. This review aims to integrate recent findings that provide insight into the molecular mechanisms underpinning astrocytic structural changes at the synapse. We offer a comprehensive discussion of the potential implications of compromised astrocyte-synapse interactions, and put forward that astrocytic synaptic coverage is generally reduced in numerous neurological disorders, with the extent of it being disease- and stage- specific. Finally, we propose outstanding questions on astrocyte-synapse structural plasticity that are relevant for future therapeutic strategies to tackle neurodegenerative and neuropsychiatric diseases.
Collapse
Affiliation(s)
- Aina Badia-Soteras
- Department of Molecular and Cellular Neuroscience, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands; Department of Brain Scienes, Imperial College London, London , United Kingdom; UK Dementia Research Institute at Imperial College London, London , United Kingdom
| | - Aline Mak
- Department of Molecular and Cellular Neuroscience, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Thomas M Blok
- Department of Molecular and Cellular Neuroscience, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Cristina Boers-Escuder
- Department of Molecular and Cellular Neuroscience, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Michel C van den Oever
- Department of Molecular and Cellular Neuroscience, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Rogier Min
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam, University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands; Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neuroscience, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Mark H G Verheijen
- Department of Molecular and Cellular Neuroscience, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands.
| |
Collapse
|
3
|
Jayathilake NJ, Phan TT, Kim J, Lee KP, Park JM. Modulating neuroplasticity for chronic pain relief: noninvasive neuromodulation as a promising approach. Exp Mol Med 2025; 57:501-514. [PMID: 40025172 PMCID: PMC11958754 DOI: 10.1038/s12276-025-01409-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/20/2024] [Accepted: 12/01/2024] [Indexed: 03/04/2025] Open
Abstract
Chronic neuropathic pain is a debilitating neuroplastic disorder that notably impacts the quality of life of millions of people worldwide. This complex condition, encompassing various manifestations, such as sciatica, diabetic neuropathy and postherpetic neuralgia, arises from nerve damage or malfunctions in pain processing pathways and involves various biological, physiological and psychological processes. Maladaptive neuroplasticity, known as central sensitization, plays a critical role in the persistence of chronic neuropathic pain. Current treatments for neuropathic pain include pharmacological interventions (for example, antidepressants and anticonvulsants), invasive procedures (for example, deep brain stimulation) and physical therapies. However, these approaches often have limitations and potential side effects. In light of these challenges, interest in noninvasive neuromodulation techniques as alternatives or complementary treatments for neuropathic pain is increasing. These methods aim to induce analgesia while reversing maladaptive plastic changes, offering potential advantages over conventional pharmacological practices and invasive methods. Recent technological advancements have spurred the exploration of noninvasive neuromodulation therapies, such as repetitive transcranial magnetic stimulation, transcranial direct current stimulation and transcranial ultrasound stimulation, as well as innovative transformations of invasive techniques into noninvasive methods at both the preclinical and clinical levels. Here this review aims to critically examine the mechanisms of maladaptive neuroplasticity in chronic neuropathic pain and evaluate the efficacy of noninvasive neuromodulation techniques in pain relief. By focusing on optimizing these techniques, we can better assess their short-term and long-term effects, refine treatment variables and ultimately improve the quality of neuropathic pain management.
Collapse
Affiliation(s)
- Nishani Jayanika Jayathilake
- Department of Physiology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Tien Thuy Phan
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Jeongsook Kim
- Department of Physiology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Kyu Pil Lee
- Department of Physiology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea.
| | - Joo Min Park
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea.
- Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
4
|
Sapienza J, Martini F, Comai S, Cavallaro R, Spangaro M, De Gregorio D, Bosia M. Psychedelics and schizophrenia: a double-edged sword. Mol Psychiatry 2025; 30:679-692. [PMID: 39294303 DOI: 10.1038/s41380-024-02743-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/20/2024]
Abstract
Psychedelics have shown promising effects in several psychiatric diseases as demonstrated by multiple clinical trials. However, no clinical experiments on patients with schizophrenia have been conducted up to date, except for some old semi-anecdotal studies mainly performed in the time-span '50s-'60s. Notably, these studies reported interesting findings, particularly on the improvement of negative symptoms and social cognition. With no doubts the lack of modern clinical studies is due to the psychomimetic properties of psychedelics, a noteworthy downside that could worsen positive symptoms. However, a rapidly increasing body of evidence has suggested that the mechanisms of action of such compounds partially overlaps with the pathogenic underpinnings of schizophrenia but in an opposite way. These findings suggest that, despite being a controversial issue, the use of psychedelics in the treatment of schizophrenia would be based on a strong biological rationale. Therefore, the aim of our perspective paper is to provide a background on the old experiments with psychedelics performed on patients with schizophrenia, interpreting them in the light of recent molecular findings on their ability to induce neuroplasticity and modulate connectivity, the immune and TAARs systems, neurotransmitters, and neurotropic factors. No systematic approach was adopted in reviewing the evidence given the difficulty to retrieve and interpret old findings. Interestingly, we identified a therapeutic potential of psychedelics in schizophrenia adopting a critical point of view, particularly on negative symptoms and social cognition, and we summarized all the relevant findings. We also identified an eligible subpopulation of chronic patients predominantly burdened by negative symptoms, outlining possible therapeutic strategies which encompass very low doses of psychedelics (microdosing), carefully considering safety and feasibility, to pave the way to future clinical trials.
Collapse
Affiliation(s)
- Jacopo Sapienza
- IRCCS San Raffaele Scientific Institute, Milan, Italy
- Department of Humanities and Life Sciences, University School for Advanced Studies IUSS, Pavia, Italy
| | | | - Stefano Comai
- IRCCS San Raffaele Scientific Institute, Milan, Italy
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Roberto Cavallaro
- IRCCS San Raffaele Scientific Institute, Milan, Italy
- School of medicine, Vita-Salute San Raffaele University, Milan, Italy
| | | | - Danilo De Gregorio
- IRCCS San Raffaele Scientific Institute, Milan, Italy
- School of medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Marta Bosia
- IRCCS San Raffaele Scientific Institute, Milan, Italy
- School of medicine, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
5
|
Zhang X, Chen Z, Xiong Y, Zhou Q, Zhu LQ, Liu D. The emerging role of nitric oxide in the synaptic dysfunction of vascular dementia. Neural Regen Res 2025; 20:402-415. [PMID: 38819044 PMCID: PMC11317957 DOI: 10.4103/nrr.nrr-d-23-01353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 10/23/2023] [Accepted: 11/30/2023] [Indexed: 06/01/2024] Open
Abstract
With an increase in global aging, the number of people affected by cerebrovascular diseases is also increasing, and the incidence of vascular dementia-closely related to cerebrovascular risk-is increasing at an epidemic rate. However, few therapeutic options exist that can markedly improve the cognitive impairment and prognosis of vascular dementia patients. Similarly in Alzheimer's disease and other neurological disorders, synaptic dysfunction is recognized as the main reason for cognitive decline. Nitric oxide is one of the ubiquitous gaseous cellular messengers involved in multiple physiological and pathological processes of the central nervous system. Recently, nitric oxide has been implicated in regulating synaptic plasticity and plays an important role in the pathogenesis of vascular dementia. This review introduces in detail the emerging role of nitric oxide in physiological and pathological states of vascular dementia and summarizes the diverse effects of nitric oxide on different aspects of synaptic dysfunction, neuroinflammation, oxidative stress, and blood-brain barrier dysfunction that underlie the progress of vascular dementia. Additionally, we propose that targeting the nitric oxide-sGC-cGMP pathway using certain specific approaches may provide a novel therapeutic strategy for vascular dementia.
Collapse
Affiliation(s)
- Xiaorong Zhang
- Department of Pathology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi Province, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi Province, China
- Center for Cognitive Science and Transdisciplinary Studies, Jiujiang University, Jiangxi Province, China
| | - Zhiying Chen
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi Province, China
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi Province, China
| | - Yinyi Xiong
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi Province, China
- Department of Rehabilitation, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi Province, China
| | - Qin Zhou
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi Province, China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Dan Liu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
6
|
Wu J, Tang J, Zhang L, Wang W, Li Z, Zhou L, Jiang X, Huang Y, Guo Q, Wang W, Ding Z, Cai F, Xi K, Gu Y, Chen L. Biomimetic "Trojan Horse" Fibers Modulate Innate Immunity Cascades for Nerve Regeneration. ACS NANO 2025; 19:781-802. [PMID: 39708371 PMCID: PMC11752508 DOI: 10.1021/acsnano.4c12036] [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: 08/29/2024] [Revised: 12/07/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Neutrophil membrane vesicles (NMVs) have been successfully applied to control the inflammatory cascade after spinal cord injury (SCI) by acting as an inflammatory factor decoy to front-load the overall inflammation regulatory window; however, the mechanisms by which NMVs regulate macrophage phenotypic shifts as well as their outcomes have rarely been reported. In this study, we demonstrated the "efferocytosis-like" effect of NMVs endocytosed by macrophages, supplementing the TCA cycle intermediate metabolite α-KG by promoting glutamine metabolism, which in turn facilitates oxidative phosphorylation and inhibits the NF-κB signaling pathway to reprogram inflammatory macrophages to the pro-regenerative phenotype. Based on these findings, a "Trojan horse" composite fiber scaffold was constructed; this comprised a carboxylated poly-l-lactic acid shell encapsulated with NMVs and a core loaded with brain-derived neurotrophic factor to spatiotemporally modulate the inflammatory microenvironment by 39.23% and sustainably promote nerve regeneration by 85.67%. In vivo experiments further confirmed the effect of NMV-coated fiber scaffolds on the regulation of early innate immune inflammation and the continuous promotion of nerve regeneration. This study not only further unravels the mechanism of neutrophil membrane-macrophage interactions but also provides a strategy for coordinating inflammatory reprogramming and nerve regeneration following SCI.
Collapse
Affiliation(s)
| | | | | | | | - Ziang Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Liang Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Xinzhao Jiang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Yiyang Huang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Qiangqiang Guo
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Wenbo Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Zhouye Ding
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Feng Cai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Kun Xi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Yong Gu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Liang Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| |
Collapse
|
7
|
Xu H, Du YN, Yang S, Guo YW. Relationship between BDNF content in cord blood and early neurobehavior in newborns with subclinical hypothyroidism during pregnancy: a preliminary study. Front Neurol 2025; 15:1465715. [PMID: 39839860 PMCID: PMC11746076 DOI: 10.3389/fneur.2024.1465715] [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: 07/17/2024] [Accepted: 12/13/2024] [Indexed: 01/23/2025] Open
Abstract
Objectives Research on neurobehavioral abnormalities in neonates of mothers with subclinical hypothyroidism (SCH) is limited. The link between umbilical cord blood brain-derived neurotrophic factor (BDNF) levels and neurobehavioral outcomes in neonates has not been explored. This study investigates the correlation between alterations in umbilical cord blood BDNF levels and early neurobehavioral abnormalities in neonates born to pregnant women with SCH. Methods This study recruited 72 pregnant women with SCH and 76 healthy controls (HC). The study collected general information for all subjects, including body mass index, parity, thyroid function assessed during early to late pregnancy, and neonatal birth weight. Neonatal behavioral and neural abilities were evaluated using the Neonatal Behavioral Neurological Assessment (NBNA). BDNF levels in umbilical cord blood were measured using the Enzyme-Linked Immunosorbent Assay method. Results The results indicated that neonates with SCH during pregnancy had lower total NBNA scores, behavioral ability, passive muscle tone, active muscle tone, primitive reflexes, general assessment, and lower levels of cord blood BDNF compared to healthy controls. The cord blood BDNF of newborns with SCH during pregnancy was positively correlated with total NBNA score, behavioral ability, active muscle tone, and general assessment. Moreover, multiple linear regression analysis demonstrated an association between cord blood BDNF levels in pregnant patients with SCH and multiple measures of newborn health, including total NBNA score, behavioral ability, active muscle tone, and general assessment. Conclusion Infants born to pregnant women with SCH exhibit reduced behavioral and neural abilities linked to BDNF levels in umbilical cord blood.
Collapse
Affiliation(s)
- Hui Xu
- Department of Obstetrics, Affiliated Maternity and Child Health Hospital of Anhui Medical University, Hefei, China
| | - Ya-Nan Du
- Department of Obstetrics, Affiliated Maternity and Child Health Hospital of Anhui Medical University, Hefei, China
| | - Shuai Yang
- Department of Neurocritical Care Unit, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yu-Wen Guo
- Department of Obstetrics, Affiliated Maternity and Child Health Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
8
|
Wang A, Huang Y, Song X, Zeng J, Zhu L, Wang B, Wu Y, Xu Z, Zheng R, Qin Y, Wang J, Yao W, Wan X, Li H, Zhuang P, Jiao J, Zhang Y, Wu Y. Parental exposure to acrylamide disrupts sphingolipid metabolism and impairs transgenerational neurodevelopment in zebrafish (Danio rerio) offspring. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 950:175134. [PMID: 39084380 DOI: 10.1016/j.scitotenv.2024.175134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
Acrylamide exposure has become an emerging environmental and food safety issue, and its toxicity poses a potential threat to public health worldwide. However, limited studies have paid attention to the detrimental effects of parental exposure to acrylamide on the neurodevelopment in zebrafish offspring. In this study, the embryos were life-cycle exposed to acrylamide (0.125 and 0.25 mM) for 180 days. Subsequently, these zebrafish (F0) were allowed to mate, and their offspring (F1) were collected to culture in clean water from embryos to adults. We employed developmental and morphological observations, behavioral profiles, metabolomics analyses, and transcriptional level examinations to investigate the transgenerational neurotoxicity with parental exposure to acrylamide. Our results showed that parental exposure to acrylamide harms the birth, development, and behavior characterization of the F1 zebrafish larvae, including poor egg quality, increased mortality rates, abnormal heart rates, slowed swimming activity, and heightened anxiety behavior, and continuously disturbs mental health in F1 adult zebrafish. The transcriptional analysis showed that parental chronic exposure to acrylamide deteriorates the neurodevelopment in F1 larvae. In addition, metabolomics analyses revealed that sphingolipid metabolism disruption may be associated with the observed abnormal development and behavioral response in unexposed F1 offspring. Overall, the present study provides pioneer evidence that acrylamide induces transgenerational neurotoxicity via targeting and disrupting sphingolipid metabolism, which reveals intergenerational transmission of acrylamide exposure and unravels its spatiotemporal toxicological effect on neurodevelopment.
Collapse
Affiliation(s)
- Anli Wang
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Yingyu Huang
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Xiaoran Song
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Jia Zeng
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Li Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Binjie Wang
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou 310053, Zhejiang, China
| | - Yuanzhao Wu
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou 310053, Zhejiang, China
| | - Zhongshi Xu
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou 310053, Zhejiang, China
| | - Ruonan Zheng
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou 310053, Zhejiang, China
| | - Yazhou Qin
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou 310053, Zhejiang, China
| | - Jiye Wang
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou 310053, Zhejiang, China
| | - Weixuan Yao
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou 310053, Zhejiang, China
| | - Xuzhi Wan
- Department of Endocrinology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Haoyu Li
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Pan Zhuang
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Jingjing Jiao
- Department of Endocrinology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Yu Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China.
| | - Yongning Wu
- NHC Key Laboratory of Food Safety Risk Assessment, Chinese Academy of Medical Sciences Research Unit (No. 2019RU014), China National Center for Food Safety Risk Assessment, Beijing 100021, China
| |
Collapse
|
9
|
Ma K, Zhang D, McDaniel K, Webb M, Newton SS, Lee FS, Qin L. A sexually dimorphic signature of activity-dependent BDNF signaling on the intrinsic excitability of pyramidal neurons in the prefrontal cortex. Front Cell Neurosci 2024; 18:1496930. [PMID: 39569070 PMCID: PMC11576208 DOI: 10.3389/fncel.2024.1496930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 10/25/2024] [Indexed: 11/22/2024] Open
Abstract
Autism spectrum disorder (ASD) is a group of neurodevelopmental disorders with strong genetic heterogeneity and more prevalent in males than females. We and others hypothesize that diminished activity-dependent neural signaling is a common molecular pathway dysregulated in ASD caused by diverse genetic mutations. Brain-derived neurotrophic factor (BDNF) is a key growth factor mediating activity-dependent neural signaling in the brain. A common single nucleotide polymorphism (SNP) in the pro-domain of the human BDNF gene that leads to a methionine (Met) substitution for valine (Val) at codon 66 (Val66Met) significantly decreases activity-dependent BDNF release without affecting basal BDNF secretion. By using mice with genetic knock-in of this human BDNF methionine (Met) allele, our previous studies have shown differential severity of autism-like social deficits in male and female BDNF+/Met mice. Pyramidal neurons are the principal neurons in the prefrontal cortex (PFC), a key brain region for social behaviors. Here, we investigated the impact of diminished activity-dependent BDNF signaling on the intrinsic excitability of pyramidal neurons in the PFC. Surprisingly, diminished activity-dependent BDNF signaling significantly increased the intrinsic excitability of pyramidal neurons in male mice, but not in female mice. Notably, significantly decreased thresholds of action potentials were observed in male BDNF+/Met mice, but not in female BDNF+/Met mice. Voltage-clamp recordings revealed that the sodium current densities were significantly increased in the pyramidal neurons of male BDNF+/Met mice, which were mediated by increased transcriptional level of Scn2a encoding sodium channel NaV 1.2. Medium after hyperpolarization (mAHP), another important parameter to determine intrinsic neuronal excitability, is strongly associated with neuronal firing frequency. Further, the amplitudes of mAHP were significantly decreased in male BDNF+/Met mice only, which were mediated by the downregulation of Kcnn2 encoding small conductance calcium-activated potassium channel 2 (SK2). This study reveals a sexually dimorphic signature of diminished activity-dependent BDNF signaling on the intrinsic neuronal excitability of pyramidal neurons in the PFC, which provides possible cellular and molecular mechanisms underpinning the sex differences in idiopathic ASD patients and human autism victims who carry BDNF Val66Met SNP.
Collapse
Affiliation(s)
- Kaijie Ma
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| | - Daoqi Zhang
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| | - Kylee McDaniel
- Department of Biotechnology, Mount Marty University, Yankton, SD, United States
| | - Maria Webb
- School of Health Sciences, University of South Dakota, Vermillion, SD, United States
| | - Samuel S. Newton
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| | - Francis S. Lee
- Department of Psychiatry, Department of Pharmacology, Sackler Institute for Developmental Psychobiology, Weill Cornell Medicine, New York, NY, United States
| | - Luye Qin
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| |
Collapse
|
10
|
Pramanik S, Devi M H, Chakrabarty S, Paylar B, Pradhan A, Thaker M, Ayyadhury S, Manavalan A, Olsson PE, Pramanik G, Heese K. Microglia signaling in health and disease - Implications in sex-specific brain development and plasticity. Neurosci Biobehav Rev 2024; 165:105834. [PMID: 39084583 DOI: 10.1016/j.neubiorev.2024.105834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/21/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
Microglia, the intrinsic neuroimmune cells residing in the central nervous system (CNS), exert a pivotal influence on brain development, homeostasis, and functionality, encompassing critical roles during both aging and pathological states. Recent advancements in comprehending brain plasticity and functions have spotlighted conspicuous variances between male and female brains, notably in neurogenesis, neuronal myelination, axon fasciculation, and synaptogenesis. Nevertheless, the precise impact of microglia on sex-specific brain cell plasticity, sculpting diverse neural network architectures and circuits, remains largely unexplored. This article seeks to unravel the present understanding of microglial involvement in brain development, plasticity, and function, with a specific emphasis on microglial signaling in brain sex polymorphism. Commencing with an overview of microglia in the CNS and their associated signaling cascades, we subsequently probe recent revelations regarding molecular signaling by microglia in sex-dependent brain developmental plasticity, functions, and diseases. Notably, C-X3-C motif chemokine receptor 1 (CX3CR1), triggering receptors expressed on myeloid cells 2 (TREM2), calcium (Ca2+), and apolipoprotein E (APOE) emerge as molecular candidates significantly contributing to sex-dependent brain development and plasticity. In conclusion, we address burgeoning inquiries surrounding microglia's pivotal role in the functional diversity of developing and aging brains, contemplating their potential implications for gender-tailored therapeutic strategies in neurodegenerative diseases.
Collapse
Affiliation(s)
- Subrata Pramanik
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| | - Harini Devi M
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Saswata Chakrabarty
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Berkay Paylar
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Ajay Pradhan
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Manisha Thaker
- Eurofins Lancaster Laboratories, Inc., 2425 New Holland Pike, Lancaster, PA 17601, USA
| | - Shamini Ayyadhury
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Arulmani Manavalan
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600077, India
| | - Per-Erik Olsson
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Gopal Pramanik
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India.
| | - Klaus Heese
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 133791, the Republic of Korea.
| |
Collapse
|
11
|
Egger K, Aicher HD, Cumming P, Scheidegger M. Neurobiological research on N,N-dimethyltryptamine (DMT) and its potentiation by monoamine oxidase (MAO) inhibition: from ayahuasca to synthetic combinations of DMT and MAO inhibitors. Cell Mol Life Sci 2024; 81:395. [PMID: 39254764 PMCID: PMC11387584 DOI: 10.1007/s00018-024-05353-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/19/2024] [Accepted: 07/04/2024] [Indexed: 09/11/2024]
Abstract
The potent hallucinogen N,N-dimethyltryptamine (DMT) has garnered significant interest in recent years due to its profound effects on consciousness and its therapeutic psychopotential. DMT is an integral (but not exclusive) psychoactive alkaloid in the Amazonian plant-based brew ayahuasca, in which admixture of several β-carboline monoamine oxidase A (MAO-A) inhibitors potentiate the activity of oral DMT, while possibly contributing in other respects to the complex psychopharmacology of ayahuasca. Irrespective of the route of administration, DMT alters perception, mood, and cognition, presumably through agonism at serotonin (5-HT) 1A/2A/2C receptors in brain, with additional actions at other receptor types possibly contributing to its overall psychoactive effects. Due to rapid first pass metabolism, DMT is nearly inactive orally, but co-administration with β-carbolines or synthetic MAO-A inhibitors (MAOIs) greatly increase its bioavailability and duration of action. The synergistic effects of DMT and MAOIs in ayahuasca or synthetic formulations may promote neuroplasticity, which presumably underlies their promising therapeutic efficacy in clinical trials for neuropsychiatric disorders, including depression, addiction, and post-traumatic stress disorder. Advances in neuroimaging techniques are elucidating the neural correlates of DMT-induced altered states of consciousness, revealing alterations in brain activity, functional connectivity, and network dynamics. In this comprehensive narrative review, we present a synthesis of current knowledge on the pharmacology and neuroscience of DMT, β-carbolines, and ayahuasca, which should inform future research aiming to harness their full therapeutic potential.
Collapse
Affiliation(s)
- Klemens Egger
- Department of Adult Psychiatry and Psychotherapy, Psychiatric University Clinic Zurich and University of Zurich, Zurich, Switzerland.
- Neuroscience Center Zurich, University of Zurich and Swiss Federal Institute of Technology Zurich, Zurich, Switzerland.
- Department of Nuclear Medicine, Bern University Hospital, Bern, Switzerland.
| | - Helena D Aicher
- Department of Adult Psychiatry and Psychotherapy, Psychiatric University Clinic Zurich and University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
- Department of Psychology, University of Zurich, Zurich, Switzerland
| | - Paul Cumming
- Department of Nuclear Medicine, Bern University Hospital, Bern, Switzerland
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, Australia
| | - Milan Scheidegger
- Department of Adult Psychiatry and Psychotherapy, Psychiatric University Clinic Zurich and University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| |
Collapse
|
12
|
Fatt MP, Zhang MD, Kupari J, Altınkök M, Yang Y, Hu Y, Svenningsson P, Ernfors P. Morphine-responsive neurons that regulate mechanical antinociception. Science 2024; 385:eado6593. [PMID: 39208104 PMCID: PMC7616448 DOI: 10.1126/science.ado6593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 07/03/2024] [Indexed: 09/04/2024]
Abstract
Opioids are widely used, effective analgesics to manage severe acute and chronic pain, although they have recently come under scrutiny because of epidemic levels of abuse. While these compounds act on numerous central and peripheral pain pathways, the neuroanatomical substrate for opioid analgesia is not fully understood. By means of single-cell transcriptomics and manipulation of morphine-responsive neurons, we have identified an ensemble of neurons in the rostral ventromedial medulla (RVM) that regulates mechanical nociception in mice. Among these, forced activation or silencing of excitatory RVMBDNF projection neurons mimicked or completely reversed morphine-induced mechanical antinociception, respectively, via a brain-derived neurotrophic factor (BDNF)/tropomyosin receptor kinase B (TrkB)-dependent mechanism and activation of inhibitory spinal galanin-positive neurons. Our results reveal a specific RVM-spinal circuit that scales mechanical nociception whose function confers the antinociceptive properties of morphine.
Collapse
Affiliation(s)
- Michael P. Fatt
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65Stockholm, Sweden
| | - Ming-Dong Zhang
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65Stockholm, Sweden
| | - Jussi Kupari
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65Stockholm, Sweden
| | - Müge Altınkök
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65Stockholm, Sweden
| | - Yunting Yang
- Division of Neuro, Department of Clinical Neuroscience, Karolinska Institutet, 171 77Stockholm, Sweden
| | - Yizhou Hu
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65Stockholm, Sweden
| | - Per Svenningsson
- Division of Neuro, Department of Clinical Neuroscience, Karolinska Institutet, 171 77Stockholm, Sweden
| | - Patrik Ernfors
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65Stockholm, Sweden
| |
Collapse
|
13
|
Singh AA, Yadav D, Khan F, Song M. Indole-3-Carbinol and Its Derivatives as Neuroprotective Modulators. Brain Sci 2024; 14:674. [PMID: 39061415 PMCID: PMC11274471 DOI: 10.3390/brainsci14070674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 06/26/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and its downstream tropomyosin receptor kinase B (TrkB) signaling pathway play pivotal roles in the resilience and action of antidepressant drugs, making them prominent targets in psychiatric research. Oxidative stress (OS) contributes to various neurological disorders, including neurodegenerative diseases, stroke, and mental illnesses, and exacerbates the aging process. The nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant responsive element (ARE) serves as the primary cellular defense mechanism against OS-induced brain damage. Thus, Nrf2 activation may confer endogenous neuroprotection against OS-related cellular damage; notably, the TrkB/phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway, stimulated by BDNF-dependent TrkB signaling, activates Nrf2 and promotes its nuclear translocation. However, insufficient neurotrophin support often leads to the downregulation of the TrkB signaling pathway in brain diseases. Thus, targeting TrkB activation and the Nrf2-ARE system is a promising therapeutic strategy for treating neurodegenerative diseases. Phytochemicals, including indole-3-carbinol (I3C) and its metabolite, diindolylmethane (DIM), exhibit neuroprotective effects through BDNF's mimetic activity; Akt phosphorylation is induced, and the antioxidant defense mechanism is activated by blocking the Nrf2-kelch-like ECH-associated protein 1 (Keap1) complex. This review emphasizes the therapeutic potential of I3C and its derivatives for concurrently activating neuronal defense mechanisms in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Alka Ashok Singh
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (A.A.S.); (D.Y.)
| | - Dhananjay Yadav
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (A.A.S.); (D.Y.)
| | - Fazlurrahman Khan
- Institute of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea;
- International Graduate Program of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea
| | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (A.A.S.); (D.Y.)
| |
Collapse
|
14
|
Ding W, Wang L, Li L, Li H, Wu J, Zhang J, Wang J. Pathogenesis of depression and the potential for traditional Chinese medicine treatment. Front Pharmacol 2024; 15:1407869. [PMID: 38983910 PMCID: PMC11231087 DOI: 10.3389/fphar.2024.1407869] [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: 03/27/2024] [Accepted: 06/05/2024] [Indexed: 07/11/2024] Open
Abstract
Depression is a prevalent mental disorder that significantly diminishes quality of life and longevity, ranking as one of the primary causes of disability globally. Contemporary research has explored the potential pathogenesis of depression from various angles, encompassing genetics, neurotransmitter systems, neurotrophic factors, the hypothalamic-pituitary-adrenal axis, inflammation, and intestinal flora, among other contributing factors. In addition, conventional chemical medications are plagued by delayed onset of action, persistent adverse effects, and restricted therapeutic efficacy. In light of these limitations, the therapeutic approach of traditional Chinese medicine (TCM) has gained increasing recognition for its superior effectiveness. Numerous pharmacological and clinical studies have substantiated TCM's capacity to mitigate depressive symptoms through diverse mechanisms. This article attempts to summarize the mechanisms involved in the pathogenesis of depression and to describe the characteristics of herbal medicines (including compounded formulas and active ingredients) for the treatment of depression. It further evaluates their effectiveness by correlating with the multifaceted pathogenesis of depression, thereby furnishing a reference for future research endeavors.
Collapse
Affiliation(s)
- Weixing Ding
- College of Traditional Chinese Medicinal Material, Jilin Agricultural University, Changchun, China
| | - Lulu Wang
- School of Medicine, Changchun Sci-Tech University, Changchun, China
| | - Lei Li
- College of Traditional Chinese Medicinal Material, Jilin Agricultural University, Changchun, China
| | - Hongyan Li
- College of Traditional Chinese Medicinal Material, Jilin Agricultural University, Changchun, China
| | - Jianfa Wu
- College of Traditional Chinese Medicinal Material, Jilin Agricultural University, Changchun, China
| | - Jing Zhang
- College of Traditional Chinese Medicinal Material, Jilin Agricultural University, Changchun, China
- Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Changchun, China
| | - Jing Wang
- Jilin Province Faw General Hospital, Changchun, China
| |
Collapse
|
15
|
Guo J, Cao Y, Zhang T, Xu C, Liu Z, Li W, Wang Q. Multisensory Fusion Training and 7, 8-Dihydroxyflavone Improve Amyloid-β-Induced Cognitive Impairment, Anxiety, and Depression-Like Behavior in Mice Through Multiple Mechanisms. Neuropsychiatr Dis Treat 2024; 20:1247-1270. [PMID: 38883414 PMCID: PMC11180438 DOI: 10.2147/ndt.s459891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/05/2024] [Indexed: 06/18/2024] Open
Abstract
Background There is growing interest in the role of physical activity in patients with of Alzheimer's disease (AD), particularly regarding its impact of cognitive function, gut microbiota, metabolites, and neurotrophic factors. Objective To investigate the impact of multisensory fusion training (MSFT) combined with 7, 8-dihydroxyflavone (DHF) on the behavioral characteristics, protein expression, microbiome, and serum metabolome using the AD model in mice induced with amyloid-β (Aβ). Methods We assessed cognitive ability, anxiety-like and depression-like behaviors in Aβ mice using behavioral measures. Western blotting was employed to detect the expression of relevant proteins. The 16S rRNA gene sequencing and metabolomics were used to analyze changes in the intestinal microbial composition and serum metabolic profile, respectively, of Aβ mice. Results The behavioral outcomes indicated that a 4-week intervention combining DHF and MSFT yielded remarkable improvements in cognitive function and reduced anxiety and depression-like behaviors in Aβ mice. In the hippocampus of Aβ mice, the combined intervention increased the levels of BDNF, VGF, PSD-95, Nrf2, p-GSK3β and p-CREB proteins. Analyses of sequence and metabolomic data revealed that Bacteroides and Ruminococcaceae were remarkably more abundant following the combined intervention, influencing the expression of specific metabolites directly linked to the maintenance of neuronal and neurobehavioral functions. These metabolites play a crucial role in vital processes, such as amino acid metabolism, lipid metabolism, and neurotransmitter metabolism in mice. Conclusion Our study highlighted that MSFT combined with DHF improves cognitive impairment, anxiety, and depression-like behavior in Aβ mice through multiple mechanisms, and further validated the correlation between the gut microbiome and serum metabolome. These findings open up a promising avenue for future investigations into potential treatment strategies for AD.
Collapse
Affiliation(s)
- Jiejie Guo
- Zhejiang Key Laboratory of Pathophysiology, NBU Health Science Center, Ningbo University, Ningbo, People's Republic of China
- Department of Clinical Laboratory, The First People's Hospital of Wenling, Taizhou, People's Republic of China
- Department of Neurology, The First People's Hospital of Wenling, Taizhou, People's Republic of China
| | - Yanzi Cao
- Zhejiang Key Laboratory of Pathophysiology, NBU Health Science Center, Ningbo University, Ningbo, People's Republic of China
| | - Ting Zhang
- Department of Clinical Laboratory, The First People's Hospital of Wenling, Taizhou, People's Republic of China
| | - Chunshuang Xu
- Zhejiang Key Laboratory of Pathophysiology, NBU Health Science Center, Ningbo University, Ningbo, People's Republic of China
| | - Zhitao Liu
- Zhejiang Key Laboratory of Pathophysiology, NBU Health Science Center, Ningbo University, Ningbo, People's Republic of China
- Fujian Normal University, Fuzhou, People's Republic of China
| | - Wanyi Li
- Zhejiang Key Laboratory of Pathophysiology, NBU Health Science Center, Ningbo University, Ningbo, People's Republic of China
| | - Qinwen Wang
- Zhejiang Key Laboratory of Pathophysiology, NBU Health Science Center, Ningbo University, Ningbo, People's Republic of China
| |
Collapse
|
16
|
Zhao X, Xiao H, Li X, Zhu L, Peng Y, Chen H, Chen L, Xu D, Wang H. Multi-organ developmental toxicity and its characteristics in fetal mice induced by dexamethasone at different doses, stages, and courses during pregnancy. Arch Toxicol 2024; 98:1891-1908. [PMID: 38522057 DOI: 10.1007/s00204-024-03707-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/14/2024] [Indexed: 03/25/2024]
Abstract
Dexamethasone is widely used in pregnant women at risk of preterm birth to reduce the occurrence of neonatal respiratory distress syndrome and subsequently reduce neonatal mortality. Studies have suggested that dexamethasone has developmental toxicity, but there is a notable absence of systematic investigations about its characteristics. In this study, we examined the effects of prenatal dexamethasone exposure (PDE) on mother/fetal mice at different doses (0.2, 0.4, or 0.8 mg/kg b.i.d), stages (gestational day 14-15 or 16-17) and courses (single- or double-course) based on the clinical practice. Results showed that PDE increased intrauterine growth retardation rate, and disordered the serum glucose, lipid and cholesterol metabolic phenotypes, and sex hormone level of mother/fetal mice. PDE was further discovered to interfere with the development of fetal lung, hippocampus and bone, inhibits steroid synthesis in adrenal and testis, and promotes steroid synthesis in the ovary and lipid synthesis in the liver, with significant effects observed at high dose, early stage and double course. The order of severity might be: ovary > lung > hippocampus/bone > others. Correlation analysis revealed that the decreased serum corticosterone and insulin-like growth factor 1 (IGF1) levels were closely related to PDE-induced low birth weight and abnormal multi-organ development in offspring. In conclusion, this study systematically confirmed PDE-induced multi-organ developmental toxicity, elucidated its characteristics, and proposed the potential "glucocorticoid (GC)-IGF1" axis programming mechanism. This research provided an experimental foundation for a comprehensive understanding of the effect and characteristics of dexamethasone on fetal multi-organ development, thereby guiding the application of "precision medicine" during pregnancy.
Collapse
Affiliation(s)
- Xiaoqi Zhao
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Hao Xiao
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Xiaomin Li
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Lu Zhu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Yu Peng
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Huijun Chen
- Department of Gynaecology and Obstetrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Liaobin Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Dan Xu
- Department of Pharmaceutical Sciences, School of Pharmacy, Wuhan University, Wuhan, 430071, China.
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China.
- Department of Gynaecology and Obstetrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China.
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
17
|
Xiang X, Xia S, Li S, Zeng Y, Wang L, Zhou Y. Study on the role and mechanism of Tan IIA in Alzheimer's disease based on CREB-BDNF-TrkB pathway. Neurosci Lett 2024; 830:137769. [PMID: 38616003 DOI: 10.1016/j.neulet.2024.137769] [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: 12/27/2023] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 04/16/2024]
Abstract
The occurrence and development of Alzheimer's disease (AD) is closely related to neuronal loss, inflammatory response, cholinergic imbalance, and Tau protein hyperphosphorylation. Previous studies have confirmed that Streptozotocin (STZ) can be used to establish a rat model of AD by injecting it into the rat brain via the lateral ventricle. Our previous research showed that Danshentone IIA (Tan IIA) can improve cognitive dysfunction in rats caused by CC chemokine ligand 2, and network pharmacology results show that Tan IIA is very likely to improve AD symptoms through the cyclic adenosine monophosphate response element binding protein (CREB), brain-derived neurotrophic factor (BDNF), and tyrosine kinase receptor protein (TrkB) pathway. The results of the water maze experiment showed that after Tan IIA treatment, the escape latency of AD rats was shortened and the number of platform crossings increased; in the new object recognition experiment, the discrimination index of AD rats significantly increased after treatment; Nissl staining and Tunel staining results showed that Tan IIA increased the number of surviving neurons in the hippocampus of cognitively impaired rats and reduced neuronal apoptosis; Bielschowsky silver staining results showed that Tan IIA reduced neurofibrillary tangles (NFTs) in the AD rats; Tan IIA can reduce the inflammatory response and oxidative stress reaction in the hippocampus of AD rats, and at the same time reduce the activity of acetylcholinesterase. Tan IIA can significantly increase the expression of CREB, BDNF, TrkB in the hippocampal tissue of STZ-injured rats (P < 0.05). These data suggest that Tan IIA may upregulate the expression of the CREB-BDNF-TrkB signaling pathway in the hippocampus of brain tissue, produce anti-neuroinflammatory, antioxidant stress, inhibit neuronal apoptosis effects, and improve cholinergic neurotransmitter disorder induced by STZ, reduce the neuronal damage and learning and memory impairment caused by STZ in rats, and improve the cognitive function of rats.
Collapse
Affiliation(s)
- Xiyong Xiang
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Siyu Xia
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Shan Li
- College of Nursing, Guangxi Medical University, Nanning 530021, China
| | - Yirong Zeng
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Lixuan Wang
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Yan Zhou
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, Guangxi Medical University, Nanning 530021, China.
| |
Collapse
|
18
|
Komori T, Okamura K, Ikehara M, Yamamuro K, Endo N, Okumura K, Yamauchi T, Ikawa D, Ouji-Sageshima N, Toritsuka M, Takada R, Kayashima Y, Ishida R, Mori Y, Kamikawa K, Noriyama Y, Nishi Y, Ito T, Saito Y, Nishi M, Kishimoto T, Tanaka KF, Hiroi N, Makinodan M. Brain-derived neurotrophic factor from microglia regulates neuronal development in the medial prefrontal cortex and its associated social behavior. Mol Psychiatry 2024; 29:1338-1349. [PMID: 38243072 PMCID: PMC11189755 DOI: 10.1038/s41380-024-02413-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 12/22/2023] [Accepted: 01/04/2024] [Indexed: 01/21/2024]
Abstract
Microglia and brain-derived neurotrophic factor (BDNF) are essential for the neuroplasticity that characterizes critical developmental periods. The experience-dependent development of social behaviors-associated with the medial prefrontal cortex (mPFC)-has a critical period during the juvenile period in mice. However, whether microglia and BDNF affect social development remains unclear. Herein, we aimed to elucidate the effects of microglia-derived BDNF on social behaviors and mPFC development. Mice that underwent social isolation during p21-p35 had increased Bdnf in the microglia accompanied by reduced adulthood sociability. Additionally, transgenic mice overexpressing microglial Bdnf-regulated using doxycycline at different time points-underwent behavioral, electrophysiological, and gene expression analyses. In these mice, long-term overexpression of microglial BDNF impaired sociability and excessive mPFC inhibitory neuronal circuit activity. However, administering doxycycline to normalize BDNF from p21 normalized sociability and electrophysiological function in the mPFC, whereas normalizing BDNF from later ages (p45-p50) did not normalize electrophysiological abnormalities in the mPFC, despite the improved sociability. To evaluate the possible role of BDNF in human sociability, we analyzed the relationship between adverse childhood experiences and BDNF expression in human macrophages, a possible proxy for microglia. Results show that adverse childhood experiences positively correlated with BDNF expression in M2 but not M1 macrophages. In summary, our study demonstrated the influence of microglial BDNF on the development of experience-dependent social behaviors in mice, emphasizing its specific impact on the maturation of mPFC function, particularly during the juvenile period. Furthermore, our results propose a translational implication by suggesting a potential link between BDNF secretion from macrophages and childhood experiences in humans.
Collapse
Affiliation(s)
- Takashi Komori
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Kazuya Okamura
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Minobu Ikehara
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Kazuhiko Yamamuro
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Nozomi Endo
- Department of Anatomy and Cell Biology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Kazuki Okumura
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Takahira Yamauchi
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Daisuke Ikawa
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | | | - Michihiro Toritsuka
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Ryohei Takada
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Yoshinori Kayashima
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Rio Ishida
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Yuki Mori
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Kohei Kamikawa
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Yuki Noriyama
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Yuki Nishi
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Toshihiro Ito
- Department of Immunology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Yasuhiko Saito
- Department of Neurophysiology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Mayumi Nishi
- Department of Anatomy and Cell Biology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Toshifumi Kishimoto
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Noboru Hiroi
- Department of Pharmacology, UT Health San Antonio, San Antonio, TX, 78229, USA
- Department of Cellular and Integrative Physiology, UT Health San Antonio, San Antonio, TX, 78229, USA
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, 78229, USA
| | - Manabu Makinodan
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan.
| |
Collapse
|
19
|
Shafiee A, Beiky M, Mohammadi I, Rajai S, Jafarabady K, Moradi S, Beikmohamadi M, Teixeira AL. Effect of smoking on Brain-Derived Neurotrophic Factor (BDNF) blood levels: A systematic review and meta-analysis. J Affect Disord 2024; 349:525-533. [PMID: 38199418 DOI: 10.1016/j.jad.2024.01.082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 12/29/2023] [Accepted: 01/04/2024] [Indexed: 01/12/2024]
Abstract
BACKGROUND Brain-Derived Neurotrophic Factor (BDNF) is a neurotrophin that plays a crucial role in neuronal survival and plasticity. Previous studies have suggested that smoking may influence BDNF levels, but the findings have been inconsistent. METHODS A comprehensive search of electronic databases was conducted to identify relevant studies. Inclusion criteria were applied to select studies that investigated the relationship between smoking and blood levels of BDNF. A random-effects model was used to estimate the overall effect size. RESULTS A total of 23 studies were included. The meta-analysis revealed a significant association between smoking and increased blood levels of BDNF (standardized mean difference [SMD] = -0.38, 95 % confidence interval [CI] 0.15 to 0.62, p = 0.002). Subgroup analyses based on BDNF source showed a significant increase in plasma-derived BDNF levels (SMD = 1.02, 95 % CI 0.50 to 1.53, p = 0.0001), while no significant difference was observed in serum-derived BDNF levels (SMD = 0.02, 95 % CI -0.19 to 0.22, p = 0.87). The pooled analysis revealed a non-significant difference in blood levels of BDNF between former smokers and non-smokers (random-effects model, SMD = 0.21, 95 % CI -0.04 to 0.46, p = 0.1). CONCLUSION Smokers exhibited significantly higher plasma levels of BDNF compared to non-smokers. Further research is needed to elucidate the underlying mechanisms and explore the potential therapeutic implications of targeting BDNF in smoking.
Collapse
Affiliation(s)
- Arman Shafiee
- Department of Psychiatry and Mental Health, Alborz University of Medical Sciences, Karaj, Iran; Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| | - Maryam Beiky
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Ida Mohammadi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahryar Rajai
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kyana Jafarabady
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Shayan Moradi
- School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahya Beikmohamadi
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Antonio Lucio Teixeira
- Neuropsychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
20
|
Abstract
It has been more than a century since Pío del Río-Hortega first characterized microglia in histological stains of brain tissue. Since then, significant advances have been made in understanding the role of these resident central nervous system (CNS) macrophages. In particular, it is now known that microglia can sense neural activity and modulate neuronal circuits accordingly. We review the mechanisms by which microglia detect changes in neural activity to then modulate synapse numbers in the developing and mature CNS. This includes responses to both spontaneous and experience-driven neural activity. We further discuss activity-dependent mechanisms by which microglia regulate synaptic function and neural circuit excitability. Together, our discussion provides a comprehensive review of the activity-dependent functions of microglia within neural circuits in the healthy CNS, and highlights exciting new open questions related to understanding more fully microglia as key components and regulators of neural circuits.
Collapse
Affiliation(s)
- Violeta Durán Laforet
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
21
|
Sun R, Xi K, Ji S, Song X, Xi D, Yin W, Shao Y, Gu W, Jiang J. TDP-43 was Involved in Radiation-induced Neuronal Damage and May Not Through the BDNF/TrkB Pathway. Radiat Res 2024; 201:240-251. [PMID: 38235539 DOI: 10.1667/rade-23-00168.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 01/05/2024] [Indexed: 01/19/2024]
Abstract
Cognitive dysfunction is the most common form of radiation-induced brain injury. TDP-43 is known to be associated with hippocampal degeneration and cognitive dysfunction, in this study we wanted to know if it also had an effect on radiation-induced hippocampus damage. At first, we found the expression of TDP-43 and p-TDP-43 was increased in the hippocampus of rats with radiation-induced cognitive dysfunction. Single-cell RNA-seq analysis of the rat hippocampus showed that TDP-43 was expressed in all cell types and was significantly upregulated in neuron cells after irradiation. Enrichment analysis of gene ontology (GO) functions and KEGG pathways showed that the differential expression genes in neuron after irradiation may be involved in synaptic plasticity. In vitro, the expression of TDP-43 was also increased in neuron cells after irradiation, while the expression of brain-derived neurotrophic factor (BDNF), TrkB, typical synaptic signature proteins (SYN, GAP43 and PSD95), β-tubulin and dendritic spines were decreased. In the irradiated neurons, the β-tubulin, dendritic and spines typical synaptic signature proteins had more severe damage in pcDNA3.1-TDP-43 plasmid transfections group, however, the damages were alleviated in the siRNA-TDP-43 plasmid transfections group. BDNF was highly expressed in the irradiated pcDNA3.1-TDP-43 plasmid transfections group, while its expression was decreased in the siRNA-TDP-43 group. The TrkB expression was significantly reduced in neurons after exposure to ionizing radiation, however, there was no significant correlation with TDP-43 expression. These data indicate that TDP-43 is involved in radiation-induced neuronal synaptic plasticity decline and developmental damage, furthermore, the BDNF/TrkB signaling pathway may not be involved in this process.
Collapse
Affiliation(s)
- Rui Sun
- Department of Oncology Radiotherapy
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Kedi Xi
- Department of Oncology Radiotherapy
| | - Shengjun Ji
- Department of Radiotherapy and Oncology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | | | - Dan Xi
- Department of Oncology Radiotherapy
| | | | | | | | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu, China
- Institute of Cell Therapy, Soochow University, Changzhou, Jiangsu, China
| |
Collapse
|
22
|
AlRuwaili R, Al-Kuraishy HM, Al-Gareeb AI, Ali NH, Alexiou A, Papadakis M, Saad HM, Batiha GES. The Possible Role of Brain-derived Neurotrophic Factor in Epilepsy. Neurochem Res 2024; 49:533-547. [PMID: 38006577 PMCID: PMC10884085 DOI: 10.1007/s11064-023-04064-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/11/2023] [Accepted: 11/14/2023] [Indexed: 11/27/2023]
Abstract
Epilepsy is a neurological disease characterized by repeated seizures. Despite of that the brain-derived neurotrophic factor (BDNF) is implicated in the pathogenesis of epileptogenesis and epilepsy, BDNF may have a neuroprotective effect against epilepsy. Thus, the goal of the present review was to highlight the protective and detrimental roles of BDNF in epilepsy. In this review, we also try to find the relation of BDNF with other signaling pathways and cellular processes including autophagy, mTOR pathway, progranulin (PGN), and α-Synuclein (α-Syn) which negatively and positively regulate BDNF/tyrosine kinase receptor B (TrkB) signaling pathway. Therefore, the assessment of BDNF levels in epilepsy should be related to other neuronal signaling pathways and types of epilepsy in both preclinical and clinical studies. In conclusion, there is a strong controversy concerning the potential role of BDNF in epilepsy. Therefore, preclinical, molecular, and clinical studies are warranted in this regard.
Collapse
Affiliation(s)
- Raed AlRuwaili
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, P.O. Box 14132, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, P.O. Box 14132, Baghdad, Iraq
| | - Naif H Ali
- Department of Internal Medicine, Medical College, Najran University, Najran, Saudi Arabia
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India
- Department of Research & Development, Funogen, Athens, Greece
- Department of Research & Development, AFNP Med, Wien, 1030, Austria
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Matrouh, 51744, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira, 22511, Egypt.
| |
Collapse
|
23
|
Bimbi G, Tongiorgi E. Chemical LTP induces confinement of BDNF mRNA under dendritic spines and BDNF protein accumulation inside the spines. Front Mol Neurosci 2024; 17:1348445. [PMID: 38450041 PMCID: PMC10914971 DOI: 10.3389/fnmol.2024.1348445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 02/05/2024] [Indexed: 03/08/2024] Open
Abstract
The neurotrophin brain-derived neurotrophic factor (BDNF) plays a key role in neuronal development and synaptic plasticity. The discovery that BDNF mRNA can be transported in neuronal dendrites in an activity-dependent manner has suggested that its local translation may support synapse maturation and plasticity. However, a clear demonstration that BDNF mRNA is locally transported and translated at activated synapses in response to long-term potentiation (LTP) is still lacking. Here, we study the dynamics of BDNF mRNA dendritic trafficking following the induction of chemical LTP (cLTP). Dendritic transport of BDNF transcripts was analyzed using the MS2 system for mRNA visualization, and chimeric BDNF-GFP constructs were used to monitor protein synthesis in living neurons. We found that within 15 min from cLTP induction, most BDNF mRNA granules become stationary and transiently accumulate in the dendritic shaft at the base of the dendritic spines, while at 30 min they accumulate inside the spine, similar to the control CamkIIα mRNA which also increased inside the spines at 60 min post-cLTP. At 60 min but not at 15 min from cLTP induction, we observed an increase in BDNF protein levels within the spines. Taken together, these findings suggest that BDNF mRNA trafficking is arrested in the early phase of cLTP, providing a local source of mRNA for BDNF translation at the base of the spine followed by translocation of both the BDNF mRNA and protein within the spine head in the late phase of LTP.
Collapse
Affiliation(s)
| | - Enrico Tongiorgi
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
24
|
Wan J, Ma L, Jiao X, Dong W, Lin J, Qiu Y, Wu W, Liu Q, Chen C, Huang H, Li S, Zheng H, Wu Y. Impaired synaptic plasticity and decreased excitability of hippocampal glutamatergic neurons mediated by BDNF downregulation contribute to cognitive dysfunction in mice induced by repeated neonatal exposure to ketamine. CNS Neurosci Ther 2024; 30:e14604. [PMID: 38332635 PMCID: PMC10853651 DOI: 10.1111/cns.14604] [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: 11/05/2023] [Revised: 12/22/2023] [Accepted: 01/03/2024] [Indexed: 02/10/2024] Open
Abstract
AIM Repeated exposure to ketamine during the neonatal period in mice leads to cognitive impairments in adulthood. These impairments are likely caused by synaptic plasticity and excitability damage. We investigated the precise role of brain-derived neurotrophic factor (BDNF) in the cognitive impairments induced by repeated ketamine exposure during the neonatal period. METHODS We evaluated the cognitive function of mice using the Morris water maze test and novel object recognition test. Western blotting and immunofluorescence were used to detect the protein levels of BDNF. Western blotting, Golgi-Cox staining, transmission electron microscopy, and long-term potentiation (LTP) recordings were used to assess synaptic plasticity in the hippocampus. The excitability of neurons was evaluated using c-Fos. In the intervention experiment, pAdeno-CaMKIIα-BDNF-mNeuronGreen was injected into the hippocampal CA1 region of mice to increase the level of BDNF. The excitability of neurons was enhanced using a chemogenetic approach. RESULTS Our findings suggest that cognitive impairments in mice repeatedly exposed to ketamine during the neonatal period are associated with downregulated BDNF protein level, synaptic plasticity damage, and decreased excitability of glutamatergic neurons in the hippocampal CA1 region. Furthermore, the specific upregulation of BDNF in glutamatergic neurons of the hippocampal CA1 region and the enhancement of excitability can improve impaired synaptic plasticity and cognitive function in mice. CONCLUSION BDNF downregulation mediates synaptic plasticity and excitability damage, leading to cognitive impairments in adulthood following repeated ketamine exposure during the neonatal period.
Collapse
Affiliation(s)
- Jie Wan
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Linhui Ma
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xinhao Jiao
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Wei Dong
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Jiatao Lin
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Yongkang Qiu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Weifeng Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Qiang Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Chen Chen
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - He Huang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Shuai Li
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Hui Zheng
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yuqing Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| |
Collapse
|
25
|
Sheethal S, Ratheesh M, Jose SP, Sandya S, Samuel S, Madhavan J. Anti-insomnia Effect of a Polyherbal Formulation on P-chlorophenyalanine Induced Experimental Animal Model. Neurochem Res 2024; 49:327-337. [PMID: 37768468 DOI: 10.1007/s11064-023-04035-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/16/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023]
Abstract
Sleep is a dynamic and controlled set of physiological and behavioural practices during which the stabilisation and restoration processes of the body take place properly. Therefore, sleep disorders, especially chronic insomnia, can harm an individual's physical and mental health. However, the therapeutic alternatives are limited and possess severe side effects. Thus, in this study, we aimed to investigate the anti-insomnia effect of a polyherbal formulation (Sleep) (SLP) on p-chlorophenyalanine (PCPA) induced insomnia in rats. Intraperitoneal injection of PCPA induced the experimental condition, and the therapeutic effect of SLP was evaluated by studying the sleep pattern and expression of various neurotransmitters and receptors, along with neurotrophins. Moreover, insomnia-associated oxidative stress and inflammation were also studied. From the findings, we found that the SLP-supplemented animals improved their sleeping behaviour and that the major neurotransmitters, hormones, and receptors were maintained at an equilibrium level. Furthermore, the neurotrophin level was increased and pro-inflammatory cytokines were reduced. The evaluation of oxidative stress markers shows that the antioxidants were significantly boosted, and as a result, lipid peroxidation was prevented. The overall findings suggest that SLP can be used as an effective medication for the treatment of sleep disorders like insomnia as it triggers the major neurotransmitter system.
Collapse
Affiliation(s)
- S Sheethal
- Department of Biochemistry, St. Thomas College, Palai, Kottayam, Kerala, 686574, India
| | - M Ratheesh
- Department of Biochemistry, St. Thomas College, Palai, Kottayam, Kerala, 686574, India.
| | - Svenia P Jose
- Department of Biochemistry, St. Thomas College, Palai, Kottayam, Kerala, 686574, India
| | - S Sandya
- -Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, Karnataka, India
| | - Sarala Samuel
- -Research and Development, Kerala Ayurveda Ltd, Athani, Ernakulam, Kerala, India
| | | |
Collapse
|
26
|
Riggs LM, Pereira EFR, Thompson SM, Gould TD. cAMP-dependent protein kinase signaling is required for ( 2R,6R)-hydroxynorketamine to potentiate hippocampal glutamatergic transmission. J Neurophysiol 2024; 131:64-74. [PMID: 38050689 PMCID: PMC11286304 DOI: 10.1152/jn.00326.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/10/2023] [Accepted: 11/23/2023] [Indexed: 12/06/2023] Open
Abstract
(2R,6R)-Hydroxynorketamine (HNK) is a ketamine metabolite that shows rapid antidepressant-like effects in preclinical studies and lacks the adverse N-methyl-d-aspartate receptor (NMDAR) inhibition-related properties of ketamine. Investigating how (2R,6R)-HNK exerts its antidepressant actions may be informative in the design of novel pharmacotherapies with improved safety and efficacy. We sought to identify the molecular substrates through which (2R,6R)-HNK induces functional changes at excitatory synapses, a prevailing hypothesis for how rapid antidepressant effects are initiated. We recorded excitatory postsynaptic potentials in hippocampal slices from male Wistar Kyoto rats, which have impaired hippocampal plasticity and are resistant to traditional antidepressants. (2R,6R)-HNK (10 µM) led to a rapid potentiation of electrically evoked excitatory postsynaptic potentials at Schaffer collateral CA1 stratum radiatum synapses. This potentiation was associated with a decrease in paired pulse facilitation, suggesting an increase in the probability of glutamate release. The (2R,6R)-HNK-induced potentiation was blocked by inhibiting either cyclic adenosine monophosphate (cAMP) or its downstream target, cAMP-dependent protein kinase (PKA). As cAMP is a potent regulator of brain-derived neurotrophic factor (BDNF) release, we assessed whether (2R,6R)-HNK exerts this acute potentiation through a rapid increase in cAMP-dependent BDNF-TrkB signaling. We found that the cAMP-PKA-dependent potentiation was not dependent on TrkB activation by BDNF, which functionally delimits the acute synaptic effects of (2R,6R)-HNK from its sustained BDNF-dependent actions in vivo. These results suggest that, by potentiating glutamate release via cAMP-PKA signaling, (2R,6R)-HNK initiates acute adaptations in fast excitatory synaptic transmission that promote structural plasticity leading to maintained antidepressant action.NEW & NOTEWORTHY Ketamine is a rapid-acting antidepressant and its preclinical effects are mimicked by its (2R,6R)-(HNK) metabolite. We found that (2R,6R)-HNK initiates acute adaptations in fast excitatory synaptic transmission by potentiating glutamate release via cAMP-PKA signaling at hippocampal Schaffer collateral synapses. This cAMP-PKA-dependent potentiation was not dependent on TrkB activation by BDNF, which functionally delimits the rapid synaptic effects of (2R,6R)-HNK from its sustained BDNF-dependent actions that are thought to maintain antidepressant action in vivo.
Collapse
Affiliation(s)
- Lace M Riggs
- Program in Neuroscience and Training Program in Integrative Membrane Biology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Edna F R Pereira
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Scott M Thompson
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Todd D Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Veterans Affairs Maryland Health Care System, Baltimore, Maryland, United States
| |
Collapse
|
27
|
Tu W, Zhang T, Li C, Jia W, Yao Z, Yi S, Chen H, Liu Y, Zhou D, Wang C, Zhang R, Shi Z, Yuan T, Zhao B, Wei L. The α 1 adrenoceptor antagonist prazosin potentiates morphine induced conditioned place preference in rats. Brain Res 2023; 1821:148614. [PMID: 37783262 DOI: 10.1016/j.brainres.2023.148614] [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: 06/10/2023] [Revised: 08/31/2023] [Accepted: 09/29/2023] [Indexed: 10/04/2023]
Abstract
The norepinephrine (NE) system is involved in pathways that regulate morphine addiction. Here, we investigated the role of α1 adrenoceptor in the ventrolateral orbital cortex (VLO) of rats with repeated morphine treatment and underlying molecular mechanisms. The rewarding properties of morphine were assessed by the conditioned place preference (CPP) paradigm. Prazosin, an α1 adrenoceptor antagonist, was microinjected into the VLO. The expression of α1 adrenoceptor, p-CaMKII/CaMKII, CRTC1, BDNF and PSD95 in the VLO were determined by immunohistochemistry or western blotting. Neurotransmitter NE in the VLO and inflammatory factors in serum were detected separately through high-performance liquid chromatography and enzyme-linked immunosorbent assay. Our experimental results showed that repeated morphine administration induced stable CPP and prazosin promoted the morphine-induced CPP. Microinjection of prazosin in the VLO not only blocked the activity of α1 adrenoceptor, decreased CaMKII phosphorylation and CRTC1, which eventually resulted in a regression of synaptic plasticity-related proteins, but also was accompanied by significantly decreasing of NE in the VLO and increasing of inflammatory cytokines in peripheral blood. These findings suggested that prazosin potentiates the addictive effects of morphine. The effect of increased CPP through reducing α1 adrenoceptor and NE was associated with the CaMKII-CRTC1 pathway and synaptic plasticity-related proteins in the VLO and inflammatory cytokines in the peripheral blood. The NE system may therefore be an underlying therapeutic target in morphine addiction. Additionally, we believe that the clinical use of prazosin in hypertensive patients with morphine abuse may be a potential risk because of its reinforcing effect on addiction.
Collapse
Affiliation(s)
- Wanyu Tu
- Xinxiang Key Laboratory of Forensic Toxicology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Tengteng Zhang
- Xinxiang Key Laboratory of Forensic Toxicology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Chenchen Li
- Xinxiang Key Laboratory of Forensic Toxicology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Wenge Jia
- Xinxiang Key Laboratory of Forensic Toxicology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Zhijun Yao
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Shanyong Yi
- Xinxiang Key Laboratory of Forensic Toxicology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Hongyun Chen
- Xinxiang Key Laboratory of Forensic Toxicology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Yuan Liu
- Xinxiang Key Laboratory of Forensic Toxicology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Danya Zhou
- Xinxiang Key Laboratory of Forensic Toxicology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Chuansheng Wang
- Henan Key Laboratory of Biological Psychiatry, the Second affiliated hospital of Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Ruiling Zhang
- Henan Key Laboratory of Biological Psychiatry, the Second affiliated hospital of Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Zhe Shi
- National Clinical Research Center for Mental Disorders, and Department of Psychaitry, the Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Tifei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Bin Zhao
- Xinxiang Key Laboratory of Forensic Toxicology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang 453003, Henan, China; Henan Key Laboratory of Biological Psychiatry, the Second affiliated hospital of Xinxiang Medical University, Xinxiang 453003, Henan, China.
| | - Lai Wei
- Xinxiang Key Laboratory of Forensic Toxicology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang 453003, Henan, China; Henan Key Laboratory of Biological Psychiatry, the Second affiliated hospital of Xinxiang Medical University, Xinxiang 453003, Henan, China.
| |
Collapse
|
28
|
Pan Y, Zong Q, Li G, Wu Z, Du T, Zhang Y, Huang Z, Ma K. Nuclear localization of alpha-synuclein induces anxiety-like behavior in mice by decreasing hippocampal neurogenesis and pathologically affecting amygdala circuits. Neurosci Lett 2023; 816:137490. [PMID: 37742940 DOI: 10.1016/j.neulet.2023.137490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/29/2023] [Accepted: 09/18/2023] [Indexed: 09/26/2023]
Abstract
Fear and anxiety are common in Parkinson's disease (PD) and may be caused by pathologies outside the dopaminergic system. Increasing evidence has shown that alpha-synuclein (α-syn) is involved in the development of anxiety in PD. In this study, we examined the effects of α-syn nuclear translocation on anxiety-like behavior in mice by overexpressing α-syn in the nuclei of the cell in the hippocampus. Our results show that α-syn overexpression in the nuclei increased the excitability of hippocampal neurons and activated NG2 glial cells and promoted the synthesis and release of γ-aminobutyric acid (GABA). And nuclear localization of α-syn led to the loss of neurotrophic factors and decreased neurogenesis. Meanwhile, the hippocampus and amygdala acted synergistically, resulting in pathologic accumulation of α-syn and gliosis in the amygdala and caused loss of interneurons. These events led to the impairments of hippocampus and amygdala function, which ultimately induced anxiety-like behavior in mice. The findings obtained in our present study indicate that excessive nuclear translocation of α-syn in hippocampal neurons and damage to the amygdala circuits may be important in the development of anxiety in PD.
Collapse
Affiliation(s)
- Yue Pan
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Qinglan Zong
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Guoxiang Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Zhengcun Wu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Tingfu Du
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Ying Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China.
| | - Zhangqiong Huang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China.
| | - Kaili Ma
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China.
| |
Collapse
|
29
|
Banushi B, Polito V. A Comprehensive Review of the Current Status of the Cellular Neurobiology of Psychedelics. BIOLOGY 2023; 12:1380. [PMID: 37997979 PMCID: PMC10669348 DOI: 10.3390/biology12111380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/26/2023] [Accepted: 10/26/2023] [Indexed: 11/25/2023]
Abstract
Psychedelic substances have gained significant attention in recent years for their potential therapeutic effects on various psychiatric disorders. This review delves into the intricate cellular neurobiology of psychedelics, emphasizing their potential therapeutic applications in addressing the global burden of mental illness. It focuses on contemporary research into the pharmacological and molecular mechanisms underlying these substances, particularly the role of 5-HT2A receptor signaling and the promotion of plasticity through the TrkB-BDNF pathway. The review also discusses how psychedelics affect various receptors and pathways and explores their potential as anti-inflammatory agents. Overall, this research represents a significant development in biomedical sciences with the potential to transform mental health treatments.
Collapse
Affiliation(s)
- Blerida Banushi
- Genetics and Genomic Medicine Department, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Vince Polito
- School of Psychological Sciences, Macquarie University, Sydney, NSW 2109, Australia;
| |
Collapse
|
30
|
Flores-Gómez GD, Apam-Castillejos DJ, Juárez-Díaz I, Fuentes-Medel E, Díaz A, Tendilla-Beltrán H, Flores G. Aripiprazole attenuates the medial prefrontal cortex morphological and biochemical alterations in rats with neonatal ventral hippocampus lesion. J Chem Neuroanat 2023; 132:102316. [PMID: 37481172 DOI: 10.1016/j.jchemneu.2023.102316] [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: 06/11/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 07/24/2023]
Abstract
Schizophrenia is a neurodevelopmental disorder characterized by a loss of dendritic spines in the medial prefrontal cortex (mPFC). Multiple subclinical and clinical studies have evidenced the ability of antipsychotics to improve neuroplasticity. In this study, it was evaluated the effect of the atypical antipsychotic aripiprazole (ARI) on the behavioral and mPFC neuronal disturbances of rats with neonatal ventral hippocampus lesion (nVHL), which is a heuristic developmental model relevant to the study of schizophrenia. ARI attenuated open field hyperlocomotion in the rats with nVHL. Also, ARI ameliorated structural neuroplasticity disturbances of the mPFC layer 3 pyramidal cells, but not in the layer 5 neurons. These effects can be associated with the ARI capability of increasing brain-derived neurotrophic factor (BDNF) levels. Moreover, in the animals with nVHL, ARI attenuated the immunoreactivity for some oxidative stress-related molecules such as the nitric oxide synthase 2 (NOS-2), 3-nitrotyrosine (3-NT), and cyclooxygenase 2 (COX-2), as well as the reactive astrogliosis in the mPFC. These results contribute to current knowledge about the neurotrophic, anti-inflammatory, and antioxidant properties of antipsychotics which may be contributing to their clinical effects and envision promising therapeutic targets for the treatment of schizophrenia.
Collapse
Affiliation(s)
| | | | - Ismael Juárez-Díaz
- Facultad de Estomatología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Estefania Fuentes-Medel
- Facultad de Ciencias Químicas (FCQ), Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Alfonso Díaz
- Facultad de Ciencias Químicas (FCQ), Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Hiram Tendilla-Beltrán
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Gonzalo Flores
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico.
| |
Collapse
|
31
|
Yao ZM, Sun XR, Huang J, Chen L, Dong SY. Astrocyte-Neuronal Communication and Its Role in Stroke. Neurochem Res 2023; 48:2996-3006. [PMID: 37329448 DOI: 10.1007/s11064-023-03966-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/03/2023] [Accepted: 06/06/2023] [Indexed: 06/19/2023]
Abstract
Astrocytes are the most abundant glial cells in the central nervous system. These cells are an important hub for intercellular communication. They participate in various pathophysiological processes, including synaptogenesis, metabolic transformation, scar production, and blood-brain barrier repair. The mechanisms and functional consequences of astrocyte-neuron signaling are more complex than previously thought. Stroke is a disease associated with neurons in which astrocytes also play an important role. Astrocytes respond to the alterations in the brain microenvironment after stroke, providing required substances to neurons. However, they can also have harmful effects. In this review, we have summarized the function of astrocytes, their association with neurons, and two paradigms of the inflammatory response, which suggest that targeting astrocytes may be an effective strategy for treating stroke.
Collapse
Affiliation(s)
- Zi-Meng Yao
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Xiao-Rong Sun
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Jie Huang
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Lei Chen
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Shu-Ying Dong
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China.
- Bengbu Medical College Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Bengbu, Anhui, China.
- Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, Anhui, China.
| |
Collapse
|
32
|
Li TT, Zhao DM, Wei YT, Li JB, Li XF, Wan Q, Zhang X, Liu XN, Yang WC, Li WZ. Effect and Mechanism of Sodium Butyrate on Neuronal Recovery and Prognosis in Diabetic Stroke. J Neuroimmune Pharmacol 2023; 18:366-382. [PMID: 37318680 DOI: 10.1007/s11481-023-10071-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 05/18/2023] [Indexed: 06/16/2023]
Abstract
Ischemic stroke is a cerebrovascular lesion caused by local ischemia and hypoxia. Diabetes mellitus (DM) is a chronic inflammatory disease that disturbs immune homeostasis and predisposes patients to ischemic stroke. The mechanism by which DM exacerbates stroke remains unclear, although it may involve disturbances in immune homeostasis. Regulatory T cells (Tregs) play a regulatory role in many diseases, but the mechanism of Tregs in diabetes complicated by stroke remains unclear. Sodium butyrate is a short-chain fatty acid that increases Treg levels. This study examined the role of sodium butyrate in the prognosis of neurological function in diabetic stroke and the mechanism by which Tregs are amplified in the bilateral cerebral hemispheres. We evaluated the brain infarct volume, observed 48-h neuronal injury and 28-day behavioral changes, and calculated the 28-day survival rate in mice. We also measured Treg levels in peripheral blood and brain tissue, recorded changes in the blood‒brain barrier and water channel proteins and neurotrophic changes in mice, measured cytokine levels and peripheral B-cell distribution in bilateral hemispheres and peripheral blood, and examined the polarization of microglia and the distribution of peripheral T-cell subpopulations in bilateral hemispheres. Diabetes significantly exacerbated the poor prognosis and neurological deficits in mice with stroke, and sodium butyrate significantly improved infarct volume, prognosis, and neurological function and showed different mechanisms in brain tissue and peripheral blood. The potential regulatory mechanism in brain tissue involved modulating Tregs/TGF-β/microglia to suppress neuroinflammation, while that in peripheral blood involved improving the systemic inflammatory response through Tregs/TGF-β/T cells.
Collapse
Affiliation(s)
- Ting-Ting Li
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Deng-Ming Zhao
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Yu-Ting Wei
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Jing-Bo Li
- The Heilongjiang Key Laboratory of Anesthesia and Intensive Care Research, Harbin Medical University, 150081, Heilongjiang Province, Harbin, China
| | - Xue-Fei Li
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Qiang Wan
- Department of Anesthesiology, The First People's Hospital of Yunnan Province, 650000, Yunnan Province, Kunming, China
| | - Xin Zhang
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Xiang-Nan Liu
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Wan-Chao Yang
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Wen-Zhi Li
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China.
- The Heilongjiang Key Laboratory of Anesthesia and Intensive Care Research, Harbin Medical University, 150081, Heilongjiang Province, Harbin, China.
| |
Collapse
|
33
|
Olczak M, Poniatowski ŁA, Siwińska A, Kwiatkowska M. Post-mortem detection of neuronal and astroglial biochemical markers in serum and urine for diagnostics of traumatic brain injury. Int J Legal Med 2023; 137:1441-1452. [PMID: 37272985 DOI: 10.1007/s00414-023-02990-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 03/21/2023] [Indexed: 06/06/2023]
Abstract
Currently available epidemiological data shows that traumatic brain injury (TBI) represents one of the leading causes of death that is associated with medico-legal practice, including forensic autopsy, criminological investigation, and neuropathological examination. Attention focused on TBI research is needed to advance its diagnostics in ante- and post-mortem cases with regard to identification and validation of novel biomarkers. Recently, several markers of neuronal, astroglial, and axonal injury have been explored in various biofluids to assess the clinical origin, progression, severity, and prognosis of TBI. Despite clinical usefulness, understanding their diagnostic accuracy could also potentially help translate them either into forensic or medico-legal practice, or both. The aim of this study was to evaluate post-mortem pro-BDNF, NSE, UCHL1, GFAP, S100B, SPTAN1, NFL, MAPT, and MBP levels in serum and urine in TBI cases. The study was performed using cases (n = 40) of fatal head injury and control cases (n = 20) of sudden death. Serum and urine were collected within ∼ 24 h after death and compared using ELISA test. In our study, we observed the elevated concentration levels of GFAP and MAPT in both serum and urine, elevated concentration levels of S100B and SPTAN1 in serum, and decreased concentration levels of pro-BDNF in serum compared to the control group. The obtained results anticipate the possible implementation of performed assays as an interesting tool for forensic and medico-legal investigations regarding TBI diagnosis where the head injury was not supposed to be the direct cause of death.
Collapse
Affiliation(s)
- Mieszko Olczak
- Department of Forensic Medicine, Center for Biostructure Research, Medical University of Warsaw, Oczki 1, 02-007, Warsaw, Poland.
| | - Łukasz A Poniatowski
- Department of Neurosurgery, Dietrich-Bonhoeffer-Klinikum, Salvador-Allende-Straße 30, 17036, Neubrandenburg, Germany
| | - Agnieszka Siwińska
- Department of Forensic Medicine, Center for Biostructure Research, Medical University of Warsaw, Oczki 1, 02-007, Warsaw, Poland
| | - Magdalena Kwiatkowska
- Department of Forensic Medicine, Center for Biostructure Research, Medical University of Warsaw, Oczki 1, 02-007, Warsaw, Poland
| |
Collapse
|
34
|
Gutiérrez-Casares JR, Segú-Vergés C, Sabate Chueca J, Pozo-Rubio T, Coma M, Montoto C, Quintero J. In silico evaluation of the role of lisdexamfetamine on attention-deficit/hyperactivity disorder common psychiatric comorbidities: mechanistic insights on binge eating disorder and depression. Front Neurosci 2023; 17:1118253. [PMID: 37457000 PMCID: PMC10347683 DOI: 10.3389/fnins.2023.1118253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a psychiatric condition well recognized in the pediatric population that can persist into adulthood. The vast majority of patients with ADHD present psychiatric comorbidities that have been suggested to share, to some extent, the pathophysiological mechanism of ADHD. Lisdexamfetamine (LDX) is a stimulant prodrug approved for treating ADHD and, in the US, also for binge eating disorder (BED). Herein, we evaluated, through a systems biology-based in silico method, the efficacy of a virtual model of LDX (vLDX) as ADHD treatment to improve five common ADHD psychiatric comorbidities in adults and children, and we explored the molecular mechanisms behind LDX's predicted efficacy. After the molecular characterization of vLDX and the comorbidities (anxiety, BED, bipolar disorder, depression, and tics disorder), we created a protein-protein interaction human network to which we applied artificial neural networks (ANN) algorithms. We also generated virtual populations of adults and children-adolescents totaling 2,600 individuals and obtained the predicted protein activity from Therapeutic Performance Mapping System models. The latter showed that ADHD molecular description shared 53% of its protein effectors with at least one studied psychiatric comorbidity. According to the ANN analysis, proteins targeted by vLDX are predicted to have a high probability of being related to BED and depression. In BED, vLDX was modeled to act upon neurotransmission and neuroplasticity regulators, and, in depression, vLDX regulated the hypothalamic-pituitary-adrenal axis, neuroinflammation, oxidative stress, and glutamatergic excitotoxicity. In conclusion, our modeling results, despite their limitations and although requiring in vitro or in vivo validation, could supplement the design of preclinical and potentially clinical studies that investigate treatment for patients with ADHD with psychiatric comorbidities, especially from a molecular point of view.
Collapse
Affiliation(s)
- José Ramón Gutiérrez-Casares
- Unidad Ambulatoria de Psiquiatría y Salud Mental de la Infancia, Niñez y Adolescencia, Hospital Perpetuo Socorro, Badajoz, Spain
| | - Cristina Segú-Vergés
- Anaxomics Biotech, Barcelona, Spain
- Research Programme on Biomedical Informatics (GRIB), Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | | | | | | | - Carmen Montoto
- Department of Medical, Takeda Farmacéutica España, Madrid, Spain
| | - Javier Quintero
- Servicio de Psiquiatría, Hospital Universitario Infanta Leonor, Departamento de Medicina Legal, Patología y Psiquiatría, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
35
|
Makinodan M, Komori T, Okamura K, Ikehara M, Yamamuro K, Endo N, Okumura K, Yamauchi T, Ikawa D, Ouji-Sageshima N, Toritsuka M, Takada R, Kayashima Y, Ishida R, Mori Y, Kamikawa K, Noriyama Y, Nishi Y, Ito T, Saito Y, Nishi M, Kishimoto T, Tanaka K, Hiroi N. Brain-derived neurotrophic factor from microglia regulates neuronal development in the medial prefrontal cortex and its associated social behavior. RESEARCH SQUARE 2023:rs.3.rs-3094335. [PMID: 37461488 PMCID: PMC10350236 DOI: 10.21203/rs.3.rs-3094335/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
Microglia and brain-derived neurotrophic factor (BDNF) are essential for the neuroplasticity that characterizes critical developmental periods. The experience-dependent development of social behaviors-associated with the medial prefrontal cortex (mPFC)-has a critical period during the juvenile period in mice. However, whether microglia and BDNF affect social development remains unclear. Herein, we aimed to elucidate the effects of microglia-derived BDNF on social behaviors and mPFC development. Mice that underwent social isolation during p21-p35 had increased Bdnf in the microglia accompanied by reduced adulthood sociability. Additionally, transgenic mice overexpressing microglia Bdnf-regulated using doxycycline at different time points-underwent behavioral, electrophysiological, and gene expression analyses. In these mice, long-term overexpression of microglia BDNF impaired sociability and excessive mPFC inhibitory neuronal circuit activity. However, administration of doxycycline to normalize BDNF from p21 normalized sociability and electrophysiological functions; this was not observed when BDNF was normalized from a later age (p45-p50). To evaluate the possible role of BDNF in human sociability, we analyzed the relationship between adverse childhood experiences and BDNF expression in human macrophages, a possible substitute for microglia. Results show that adverse childhood experiences positively correlated with BDNF expression in M2 but not M1 macrophages. Thus, microglia BDNF might regulate sociability and mPFC maturation in mice during the juvenile period. Furthermore, childhood experiences in humans may be related to BDNF secretion from macrophages.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - T Ito
- Keio University School of Medicine
| | | | | | | | | | - Noboru Hiroi
- University of Texas Health Science Center at San Antonio
| |
Collapse
|
36
|
Fang L, Li CH, Zhang Q, Jiang TJ, Liu Y, Shi FP, Yu P, Yu L, Chen AP, Li T, Wan YZ, Shi L. Ciliated Cells Express a Novel Pattern of Brain-Derived Neurotrophic Factor in Allergic Rhinitis. J Inflamm Res 2023; 16:2595-2606. [PMID: 37360625 PMCID: PMC10289300 DOI: 10.2147/jir.s407368] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 06/06/2023] [Indexed: 06/28/2023] Open
Abstract
Background Mounting research indicates that brain-derived neurotrophic factor (BDNF), has great potential to increase neuro-hyperresponsiveness and airway resistance in airway allergic disease. The expression level of BDNF has been found to be notably elevated in lung/nasal lavage (NAL) fluid. However, the expression and position of BDNF in ciliated cells with allergic rhinitis remains unclear. Methods Nasal mucosal cells were collected from patients with allergic rhinitis (AR) and mice which were performed under different allergen challenge time, then observed the expression and position of BDNF located in ciliated cells through the immunofluorescence staining. Nasal mucosa, serum and NAL fluid were collected also. The expression level of BDNF and IL-4/5/13 were detected by RT-PCR. The expressions of BDNF (in serum and NAL fluid), and total-IgE, ovalbumin sIgE (in serum) were detected by ELISA. Results We found that MFI of BDNF in AR group's ciliated cells was obviously lower than that in the control group, and a negative correlation was discovered between MFI and VAS score. It can be roughly divided into 5 patterns according to its location in the cytoplasm of ciliated cells. In the mouse model, the expressions of BDNF in serum and NAL fluid increased temporarily after allergen stimulation. The MFI of BDNF in ciliated cells displayed an initial increase followed by a subsequent decrease. Conclusion Our study shows for the first time that, the expression and localization of BNDF were observed in the human nasal ciliated epithelial cells of allergic rhinitis, and the expression of level was less than the control group under the persistent state of allergy. BDNF expression in ciliated cells was transient increased after allergen stimulation and decreased to normal level after 24h in mouse model of allergic rhinitis. This might be the possible source of the transient increase of BNDF in serum and NAL fluid.
Collapse
Affiliation(s)
- Li Fang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
- Department of Otolaryngology Head & Neck Surgery, The Second People’s Hospital of Shenzhen, Shenzhen, Guangdong, People’s Republic of China
| | - Chun-Hao Li
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
- Department of Allergy, Shandong Second Provincial General Hospital, Jinan, Shandong, People’s Republic of China
| | - Qian Zhang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
- Department of Allergy, Shandong Second Provincial General Hospital, Jinan, Shandong, People’s Republic of China
| | - Tian-Jiao Jiang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
- Department of Allergy, Shandong Second Provincial General Hospital, Jinan, Shandong, People’s Republic of China
| | - Yuan Liu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
- Department of Otolaryngology Head & Neck Surgery, The Second People’s Hospital of Shenzhen, Shenzhen, Guangdong, People’s Republic of China
| | - Feng-Po Shi
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
- Department of Allergy, Shandong Second Provincial General Hospital, Jinan, Shandong, People’s Republic of China
| | - Peng Yu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
- Department of Allergy, Shandong Second Provincial General Hospital, Jinan, Shandong, People’s Republic of China
| | - Liang Yu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
- Department of Allergy, Shandong Second Provincial General Hospital, Jinan, Shandong, People’s Republic of China
| | - Ai-Ping Chen
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
- Department of Allergy, Shandong Second Provincial General Hospital, Jinan, Shandong, People’s Republic of China
| | - Tao Li
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
- Department of Allergy, Shandong Second Provincial General Hospital, Jinan, Shandong, People’s Republic of China
| | - Yu-Zhu Wan
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
- Department of Allergy, Shandong Second Provincial General Hospital, Jinan, Shandong, People’s Republic of China
| | - Li Shi
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
- Department of Allergy, Shandong Second Provincial General Hospital, Jinan, Shandong, People’s Republic of China
| |
Collapse
|
37
|
Wang W, Li X, Qian Q, Yan J, Huang H, Wang X, Wang H. Mechanistic exploration on neurodevelopmental toxicity induced by upregulation of alkbh5 targeted by triclosan exposure to larval zebrafish. JOURNAL OF HAZARDOUS MATERIALS 2023; 457:131831. [PMID: 37320907 DOI: 10.1016/j.jhazmat.2023.131831] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/31/2023] [Accepted: 06/09/2023] [Indexed: 06/17/2023]
Abstract
Because triclosan (TCS) has been confirmed to cause severe neurotoxicity, it is urgent to disclose the underlying toxicity mechanisms at varying levels. TCS exposure resulted in a series of malformations in larval zebrafish, including reduced neurons, blood-vessel ablation and abnormal neurobehavior. Apoptosis staining and the upregulated expression of proapoptotic genes demonstrated that TCS induced neuronal apoptosis and neurotransmitter disorders. By integrating RT-qPCR analysis with the effects of pathway inhibitors and agonists, we found that TCS triggered abnormal regulation of neuron development-related functional genes, and suppressed the BDNF/TrkB signaling pathway. TCS inhibited total m6A-RNA modification level by activating the demethylase ALKBH5, and induced neurodevelopmental toxicity based on the knockdown experiments of alkbh5 and molecular docking. The main novelties of this study lies in: (1) based on specific staining and transgenic lines, the differential neurotoxicity effects of TCS were unravelled at individual, physiological, biochemical and molecular levels in vivo; (2) from a epigenetics viewpoint, the decreasing m6A methylation level was confirmed to be mediated by alkbh5 upregulation; and (3) both homology modeling and molecular docking evidenced the targeting action of TCS on ALKBH5 enzyme. These findings open a novel avene for TCS's risk assessment and early intervention of the contaminant-sourcing diseases.
Collapse
Affiliation(s)
- Weiwei Wang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China; School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Xin Li
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Qiuhui Qian
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Jin Yan
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Haishan Huang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Xuedong Wang
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China.
| | - Huili Wang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China; School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China.
| |
Collapse
|
38
|
Chang HX, Dai W, Bao JH, Li JF, Zhang JG, Li YF. Essential role of microglia in the fast antidepressant action of ketamine and hypidone hydrochloride (YL-0919). Front Pharmacol 2023; 14:1122541. [PMID: 37305539 PMCID: PMC10250639 DOI: 10.3389/fphar.2023.1122541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 05/19/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction: Intracerebral microglia play a vital role in mediating central immune response, neuronal repair and synaptic pruning, but its precise role and mechanism in fast action of antidepressants have remained unknown. In this study, we identified that the microglia contributed to the rapid action of antidepressants ketamine and YL-0919. Methods: The depletion of microglia was achieved with the diet containing the colony-stimulating factor 1 receptor (CSF1R) inhibitor PLX5622 in mice. The tail suspension test (TST), forced swimming test (FST) and novelty suppressed feeding test (NSFT) were employed to evaluate the rapid acting antidepressant behavior of ketamine and YL-0919 in the microglia depletion model. The number of microglia in the prefrontal cortex (PFC) was assayed by the immunofluorescence staining. The expressions of synaptic proteins (synapsin-1, PSD-95, GluA1) and brain-derived neurotrophic factor (BDNF) in the PFC were tested by Western blot. Results: The immobility duration in FST and the latency to feed in NSFT were shortened 24 h after an intraperitoneal (i.p.) injection of ketamine (10 mg/kg). The microglial depletion of PLX3397 blocked the rapid antidepressant-like effect of ketamine in mice. In addition, the immobility time in TST and FST as well as latency to feed in NSFT were reduced 24 h after the intragastric (i.g.) administration of YL-0919 (2.5 mg/kg), and the rapid antidepressant effect of YL-0919 was also blocked by the microglial depletion using PLX5622. About 92% of microglia in the prefrontal cortex was depleted in PLX5622 diet-fed mice, while both ketamine and YL-0919 promoted proliferation on the remaining microglia. YL-0919 significantly increased the protein expressions of synapsin-1, PSD-95, GluA1 and BDNF in the PFC, all of which could be blocked by PLX5622. Conclusion: These results suggested the microglia underlying the rapid antidepressant-like effect of ketamine and YL-0919, and microglia would likely constitute in the rapid enhancing impact of synaptic plasticity in the prefrontal cortex by YL-0919.
Collapse
Affiliation(s)
- Hai-Xia Chang
- College of Pharmacy, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Wei Dai
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratories of Neuropsychopharmacology, Institute of Pharmacology and Toxicology, Beijing, China
| | - Jin-Hao Bao
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jin-Feng Li
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Ji-Guo Zhang
- College of Pharmacy, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Yun-Feng Li
- Beijing Institute of Basic Medical Sciences, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratories of Neuropsychopharmacology, Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
39
|
Tang Y, Xu Z, Tang J, Xu Y, Li Z, Wang W, Wu L, Xi K, Gu Y, Chen L. Architecture-Engineered Electrospinning Cascade Regulates Spinal Microenvironment to Promote Nerve Regeneration. Adv Healthc Mater 2023; 12:e2202658. [PMID: 36652529 PMCID: PMC11469115 DOI: 10.1002/adhm.202202658] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/14/2023] [Indexed: 01/19/2023]
Abstract
The inflammatory cascade after spinal cord injury (SCI) causes necrotizing apoptosis of local stem cells, which limits nerve regeneration. Therefore, coordinating the inflammatory immune response and neural stem cell (NSC) functions is key to promoting the recovery of central nervous system function. In this study, a hydrogel "perfusion" system and electrospinning technology are integrated, and a "concrete" composite support for the repair of nerve injuries is built. The hydrogel's hydrophilic properties activate macrophage integrin receptors to mediate polarization into anti-inflammatory subtypes and cause a 10% increase in polarized M2 macrophages, thus reprogramming the SCI immune microenvironment. Programmed stromal cell-derived factor-1α and brain-derived neurotrophic factor released from the composite increase recruitment and neuronal differentiation of NSCs by approximately four- and twofold, respectively. The fiber system regulates the SCI immune inflammatory microenvironment, recruits endogenous NSCs, promotes local blood vessel germination and maturation, and improves nerve function recovery in a rat SCI model. In conclusion, the engineering fiber composite improves the local inflammatory response. It promotes nerve regeneration through a hydrophilic programmed cytokine-delivery system, which further improves and supplements the immune response mechanism regulated by the inherent properties of the biomaterial. The new fiber composite may serve as a new treatment approach for SCI.
Collapse
Affiliation(s)
- Yu Tang
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Zonghan Xu
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Jincheng Tang
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Yichang Xu
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Ziang Li
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Wenbo Wang
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Liang Wu
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Kun Xi
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Yong Gu
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Liang Chen
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| |
Collapse
|
40
|
Ehinger Y, Soneja D, Phamluong K, Salvi A, Ron D. Identification of Novel BDNF-Specific Corticostriatal Circuitries. eNeuro 2023; 10:ENEURO.0238-21.2023. [PMID: 37156610 PMCID: PMC10198608 DOI: 10.1523/eneuro.0238-21.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 03/23/2023] [Accepted: 04/03/2023] [Indexed: 05/10/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is released from axon terminals originating in the cerebral cortex onto striatal neurons. Here, we characterized BDNF neurons in the corticostriatal circuitry. First, we used BDNF-Cre and Ribotag transgenic mouse lines to label BDNF-positive neurons in the cortex and detected BDNF expression in all the subregions of the prefrontal cortex (PFC). Next, we used a retrograde viral tracing strategy, in combination with BDNF-Cre knock-in mice, to map the cortical outputs of BDNF neurons in the dorsomedial and dorsolateral striatum (DMS and DLS, respectively). We found that BDNF-expressing neurons located in the medial prefrontal cortex (mPFC) project mainly to the DMS, and those located in the primary and secondary motor cortices (M1 and M2, respectively) and agranular insular cortex (AI) project mainly to the DLS. In contrast, BDNF-expressing orbitofrontal cortical (OFC) neurons differentially target the dorsal striatum (DS) depending on their mediolateral and rostrocaudal location. Specifically, the DMS is mainly innervated by the medial and ventral part of the orbitofrontal cortex (MO and VO, respectively), whereas the DLS receives projections specifically from the lateral part of the OFC (LO). Together, our study uncovers previously unknown BDNF corticostriatal circuitries. These findings could have important implications for the role of BDNF signaling in corticostriatal pathways.
Collapse
Affiliation(s)
- Yann Ehinger
- Department of Neurology, University of California, San Francisco, 94143-0663 CA
| | - Drishti Soneja
- Department of Neurology, University of California, San Francisco, 94143-0663 CA
| | - Khanhky Phamluong
- Department of Neurology, University of California, San Francisco, 94143-0663 CA
| | - Alexandra Salvi
- Department of Neurology, University of California, San Francisco, 94143-0663 CA
| | - Dorit Ron
- Department of Neurology, University of California, San Francisco, 94143-0663 CA
| |
Collapse
|
41
|
Wang H, Yang Y, Pei G, Wang Z, Chen N. Neurotrophic basis to the pathogenesis of depression and phytotherapy. Front Pharmacol 2023; 14:1182666. [PMID: 37089920 PMCID: PMC10115971 DOI: 10.3389/fphar.2023.1182666] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 03/28/2023] [Indexed: 04/09/2023] Open
Abstract
Depression is a major neuropsychiatric disease that considerably impacts individuals’ psychosocial function and life quality. Neurotrophic factors are now connected to the pathogenesis of depression, while the definitive neurotrophic basis remains elusive. Besides, phytotherapy is alternative to conventional antidepressants that may minimize undesirable adverse reactions. Thus, further research into the interaction between neurotrophic factors and depression and phytochemicals that repair neurotrophic factors deficit is highly required. This review highlighted the implication of neurotrophic factors in depression, with a focus on the brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF), vascular endothelial growth factor (VEGF), and nerve growth factor (NGF), and detailed the antidepressant activities of various phytochemicals targeting neurotrophic factors. Additionally, we presented future opportunities for novel diagnostic and therapeutic strategies for depression and provided solutions to challenges in this area to accelerate the clinical translation of neurotrophic factors for the treatment of depression.
Collapse
Affiliation(s)
- Huiqin Wang
- Hunan University of Chinese Medicine and Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, Hunan, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yantao Yang
- Hunan University of Chinese Medicine and Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, Hunan, China
| | - Gang Pei
- Hunan University of Chinese Medicine and Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, Hunan, China
| | - Zhenzhen Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Naihong Chen
- Hunan University of Chinese Medicine and Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, Hunan, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Naihong Chen,
| |
Collapse
|
42
|
Tao FF, Wang ZY, Wang Y, Lv QR, Cai PP, Min HW, Ge JW, Yin CY, Cheng R. Inhibition of hippocampal cyclin-dependent kinase 5 activity ameliorates learning and memory dysfunction in a mouse model of bronchopulmonary dysplasia. CNS Neurosci Ther 2023. [PMID: 36964998 DOI: 10.1111/cns.14185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/27/2023] [Accepted: 03/05/2023] [Indexed: 03/27/2023] Open
Abstract
AIMS Oxygen therapy plays a vital role in the development of bronchopulmonary dysplasia (BPD), which is the independent risk factor for neurodevelopment deficits in premature infants. However, the effect of hippocampal cyclin-dependent kinase 5 (CDK5) on BPD-associated neurodevelopment deficits is not fully understood. METHODS Mice were placed in a hyperoxia chamber from postnatal Day 1 to Day 7. Hematoxylin and eosin staining was used to evaluate the lung histomorphological characteristics. Learning and memory functions of mice were detected by Morris water maze. TUNEL staining was applied to measure the number of apoptotic cells. The expression of CDK5, apoptosis-related protein, and neuroplasticity-related proteins were analyzed by Western blot. Golgi staining was used to assess the structure of dendritic spines. RESULTS Hyperoxia-induced BPD mice showed a long-term learning and memory dysfunction, more severe neuronal apoptosis, and a decline of synaptic plasticity. Inhibition of CDK5 overactivation ameliorated cognitive deficits, neuronal apoptosis, and synaptic plasticity disorders in BPD mice. CONCLUSIONS This study first found a vital role of CDK5 in BPD-associated neurodevelopmental disorders. Inhibition of CDK5 overexpression could effectively improve cognitive dysfunctions in BPD mice, which indicated that hippocampal CDK5 may be a new target for prevention and treatment in learning and memory dysfunction of BPD.
Collapse
Affiliation(s)
- Fang-Fei Tao
- Department of Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zi-Yu Wang
- Department of Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Ying Wang
- Department of Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Qian-Ru Lv
- Department of Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Peng-Peng Cai
- Department of Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing, China
| | | | - Jian-Wei Ge
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Chun-Yu Yin
- Department of Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Rui Cheng
- Department of Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
43
|
Islam J, Shila TT, Islam Z, Kabir E, Haque N, Khatun M, Khan S, Jubayar AM, Islam F, Nikkon F, Hossain K, Saud ZA. Clerodendrum viscosum leaves attenuate lead-induced neurotoxicity through upregulation of BDNF-Akt-Nrf2 pathway in mice. JOURNAL OF ETHNOPHARMACOLOGY 2023; 304:116024. [PMID: 36549369 DOI: 10.1016/j.jep.2022.116024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 11/20/2022] [Accepted: 12/03/2022] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Clerodendrum viscosum is an important medicinal plant in Ayurveda in Bangladesh and its leaves are used as a remedy for various diseases such as anti-inflammatory, antibacterial, hyperglycemic, hepatoprotective effects. AIM OF THE STUDY The present study aimed to evaluate the protective effect of aqueous extract of C. viscosum leaves against Pb-induced neurobehavioral and biochemical changes in mice. MATERIALS AND METHODS Swiss albino mice were divided as a) control, b) lead treated (Pb) and c) C. viscosum leaves (Cle) d) Pb plus Cle groups. Pb-acetate (10 mg/kg body weight) was given to Pb and Pb + Cle groups mice, and water extract of leaves (50 mg/kg body weight) was provided as supplementation to Cle and Pb + Cle groups mice for 30 days. Elevated plus maze and Morris water maze tests were used for evaluating anxiety, spatial memory and learning, respectively. Status of cholinesterase, SOD, GSH enzyme activity and neurotoxicity markers such BDNF and Nrf2 levels were analyzed in the brain tissue of experimental mice. RESULTS Poorer learning, inferior spatial memory, and increased anxiety-like behavior in Pb-exposure mice were noted when compared to control mice in Morris water maze and elevated plus maze test, respectively. In addition, expression of BDNF and Nrf2, cholinesterase activity along with antioxidant activity were significantly reduced compared to control group (p < 0.01). Interestingly, C. viscosum leaves' aqueous extract supplementation in Pb-exposed mice provide a significant improved neurochemical and antioxidant properties through the augmentation of activity of cholinergic enzymes, and upregulation of BDNF and Nrf2 levels in the brain tissue compared to Pb-exposed mice. CONCLUSIONS This study suggested that C. viscosum leaves restore the cognitive dysfunction and reduce anxiety-like behavior through upregulation of BDNF mediated Akt-Nrf2 pathway in Pb-exposure mice.
Collapse
Affiliation(s)
- Jahidul Islam
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh.
| | - Tasnim Tabassum Shila
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh.
| | - Zohurul Islam
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh.
| | - Ehsanul Kabir
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh.
| | - Nazmul Haque
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh.
| | - Moriom Khatun
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh.
| | - Shuchismita Khan
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh.
| | - Ahsanul Mahbub Jubayar
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh.
| | - Farhadul Islam
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh.
| | - Farjana Nikkon
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh.
| | - Khaled Hossain
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh.
| | - Zahangir Alam Saud
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh.
| |
Collapse
|
44
|
Zheng X, Chen L, Chen T, Cao M, Zhang B, Yuan C, Zhao Z, Li C, Zhou X. The Mechanisms of BDNF Promoting the Proliferation of Porcine Follicular Granulosa Cells: Role of miR-127 and Involvement of the MAPK-ERK1/2 Pathway. Animals (Basel) 2023; 13:ani13061115. [PMID: 36978655 PMCID: PMC10044701 DOI: 10.3390/ani13061115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
As a member of the neurotrophic family, brain-derived neurotrophic factor (BDNF) provides a key link in the physiological process of mammalian ovarian follicle development, in addition to its functions in the nervous system. The emphasis of this study lay in the impact of BDNF on the proliferation of porcine follicular granulosa cells (GCs) in vitro. BDNF and tyrosine kinase B (TrkB, receptor of BDNF) were detected in porcine follicular GCs. Additionally, cell viability significantly increased during the culture of porcine GCs with BDNF (100 ng/mL) in vitro. However, BDNF knockdown in GCs decreased cell viability and S-phase cells proportion-and BDNF simultaneously regulated the expression of genes linked with cell proliferation (CCND1, p21 and Bcl2) and apoptosis (Bax). Then, the results of the receptor blocking experiment showed that BDNF promoted GC proliferation via TrkB. The high-throughput sequencing showed that BDNF also regulated the expression profiles of miRNAs in GCs. The differential expression profiles were obtained by miRNA sequencing after BDNF (100 ng/mL) treatment with GCs. The sequencing results showed that, after BDNF treatment, 72 significant differentially-expressed miRNAs were detected-5 of which were related to cell process and proliferation signaling pathways confirmed by RT-PCR. Furthermore, studies showed that BDNF promoted GCs' proliferation by increasing the expression of CCND1, downregulating miR-127 and activating the ERK1/2 signal pathway. Moreover, BDNF indirectly activated the ERK1/2 signal pathway by downregulating miR-127. In conclusion, BDNF promoted porcine GC proliferation by increasing CCND1 expression, downregulating miR-127 and stimulating the MAPK-ERK1/2 signaling cascade.
Collapse
Affiliation(s)
- Xue Zheng
- Laboratory for Regulation of Reproduction, College of Animal Sciences, Jilin University, Changchun 130062, China
- College of Biological and Pharmaceutical Engineering, Jilin Agricultural Science and Technology University, Jilin 132101, China
| | - Lu Chen
- Laboratory for Regulation of Reproduction, College of Animal Sciences, Jilin University, Changchun 130062, China
| | - Tong Chen
- Laboratory for Regulation of Reproduction, College of Animal Sciences, Jilin University, Changchun 130062, China
| | - Maosheng Cao
- Laboratory for Regulation of Reproduction, College of Animal Sciences, Jilin University, Changchun 130062, China
| | - Boqi Zhang
- Laboratory for Regulation of Reproduction, College of Animal Sciences, Jilin University, Changchun 130062, China
| | - Chenfeng Yuan
- Laboratory for Regulation of Reproduction, College of Animal Sciences, Jilin University, Changchun 130062, China
| | - Zijiao Zhao
- Laboratory for Regulation of Reproduction, College of Animal Sciences, Jilin University, Changchun 130062, China
| | - Chunjin Li
- Laboratory for Regulation of Reproduction, College of Animal Sciences, Jilin University, Changchun 130062, China
| | - Xu Zhou
- Laboratory for Regulation of Reproduction, College of Animal Sciences, Jilin University, Changchun 130062, China
| |
Collapse
|
45
|
Kalinichenko SG, Pushchin II, Matveeva NY. Neurotoxic and cytoprotective mechanisms in the ischemic neocortex. J Chem Neuroanat 2023; 128:102230. [PMID: 36603664 DOI: 10.1016/j.jchemneu.2022.102230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/30/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023]
Abstract
Neuronal damage in ischemic stroke occurs due to permanent imbalance between the metabolic needs of the brain and the ability of the blood-vascular system to maintain glucose delivery and adequate gas exchange. Oxidative stress and excitotoxicity trigger complex processes of neuroinflammation, necrosis, and apoptosis of both neurons and glial cells. This review summarizes data on the structural and chemical changes in the neocortex and main cytoprotective effects induced by focal ischemic stroke. We focus on the expression of neurotrophins (NT) and molecular and cellular changes in neurovascular units in ischemic brain. We also discuss how these factors affect the apoptosis of cortical cells. Ischemic damage involves close interaction of a wide range of signaling molecules, each acting as an efficient marker of cell state in both the ischemic core and penumbra. NTs play the main regulatory role in brain tissue recovery after ischemic injury. Heterogeneous distribution of the BDNF, NT-3, and GDNF immunoreactivity is concordant with the selective response of different types of cortical neurons and glia to ischemic injury and allows mapping the position of viable neurons. Astrocytes are the central link in neurovascular coupling in ischemic brain by providing other cells with a wide range of vasotropic factors. The NT expression coincides with the distribution of reactive astrocytes, marking the boundaries of the penumbra. The development of ischemic stroke is accompanied by a dramatic change in the distribution of GDNF reactivity. In early ischemic period, it is mainly observed in cortical neurons, while in late one, the bulk of GDNF-positive cells are various types of glia, in particular, astrocytes. The proportion of GDNF-positive astrocytes increases gradually throughout the ischemic period. Some factors that exert cytoprotective effects in early ischemic period may display neurotoxic and pro-apoptotic effects later on. The number of apoptotic cells in the ischemic brain tissue correlates with the BDNF levels, corroborating its protective effects. Cytoprotection and neuroplasticity are two lines of brain protection and recovery after ischemic stroke. NTs can be considered an important link in these processes. To develop efficient pharmacological therapy for ischemic brain injury, we have to deepen our understanding of neurochemical adaptation of brain tissue to acute stroke.
Collapse
Affiliation(s)
- Sergei G Kalinichenko
- Department of Histology, Cytology, and Embryology, Pacific State Medical University, Vladivostok 690950, Russia
| | - Igor I Pushchin
- Laboratory of Physiology, A.V. Zhirmusky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia.
| | - Natalya Yu Matveeva
- Department of Histology, Cytology, and Embryology, Pacific State Medical University, Vladivostok 690950, Russia
| |
Collapse
|
46
|
Woodburn SC, Asrat HS, Flurer JK, Schwierling HC, Bollinger JL, Vollmer LL, Wohleb ES. Depletion of microglial BDNF increases susceptibility to the behavioral and synaptic effects of chronic unpredictable stress. Brain Behav Immun 2023; 109:127-138. [PMID: 36681359 PMCID: PMC10023455 DOI: 10.1016/j.bbi.2023.01.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 12/22/2022] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
In the medial prefrontal cortex (PFC), chronic stress reduces synaptic expression of glutamate receptors, leading to decreased excitatory signaling from layer V pyramidal neurons and working memory deficits. One key element driving these changes is a reduction in brain-derived neurotrophic factor (BDNF) signaling. BDNF is a potent mediator of synaptic growth and deficient BDNF signaling has been linked to stress susceptibility. Prior studies indicated that neurons are the primary source of BDNF, but more recent work suggests that microglia are also an important source of BDNF. Adding to this, our work showed that 14 days of chronic unpredictable stress (CUS) reduced Bdnf transcript in PFC microglia, evincing its relevance in the effects of stress. To explore this further, we utilized transgenic mice with microglia-specific depletion of BDNF (Cx3cr1Cre/+:Bdnffl/fl) and genotype controls (Cx3cr1Cre/+:Bdnf+/+). In the following experiments, mice were exposed to a shortened CUS paradigm (7 days) to determine if microglial Bdnf depletion promotes stress susceptibility. Analyses of PFC microglia revealed that Cx3cr1Cre/+:Bdnffl/fl mice had shifts in phenotypic markers and gene expression. In a separate cohort, synaptoneurosomes were collected from the PFC and western blotting was performed for synaptic markers. These experiments showed that Cx3cr1Cre/+:Bdnffl/fl mice had baseline deficits in GluN2B, and that 7 days of CUS additionally reduced GluN2A levels in Cx3cr1Cre/+:Bdnffl/fl mice, but not genotype controls. Behavioral and cognitive testing showed that this coincided with exacerbated stress effects on temporal object recognition in Cx3cr1Cre/+:Bdnffl/fl mice. These results indicate that microglial BDNF promotes glutamate receptor expression in the PFC. As such, mice with deficient microglial BDNF had increased susceptibility to the behavioral and cognitive consequences of stress.
Collapse
Affiliation(s)
- Samuel C Woodburn
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Helina S Asrat
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - James K Flurer
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Hana C Schwierling
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Justin L Bollinger
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Lauren L Vollmer
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Eric S Wohleb
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
47
|
Yoshimura R, Okamoto N, Chibaatar E, Natsuyama T, Ikenouchi A. The Serum Brain-Derived Neurotrophic Factor Increases in Serotonin Reuptake Inhibitor Responders Patients with First-Episode, Drug-Naïve Major Depression. Biomedicines 2023; 11:biomedicines11020584. [PMID: 36831119 PMCID: PMC9953440 DOI: 10.3390/biomedicines11020584] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a growth factor synthesized in the cell bodies of neurons and glia, which affects neuronal maturation, the survival of nervous system, and synaptic plasticity. BDNF play an important role in the pathophysiology of major depression (MD). The serum BDNF levels changed over time, or with the improvement in depressive symptoms. However, the change of serum BDNF during pharmacotherapy remains obscure in MDD. In particular, the changes in serum BDNF associated with pharmacotherapy have not yet been fully elucidated. The present study aimed to compare the changes in serum BDNF concentrations in first-episode, drug-naive patients with MD treated with antidepressants between treatment-response and treatment-nonresponse groups. The study included 35 inpatients and outpatients composed of 15 males and 20 females aged 36.7 ± 6.8 years at the Department of Psychiatry of our University Hospital. All patients met the DSM-5 diagnostic criteria for MD. The antidepressants administered included paroxetine, duloxetine, and escitalopram. Severity of depressive state was assessed using the 17-item HAMD before and 8 weeks after drug administration. Responders were defined as those whose total HAMD scores at 8 weeks had decreased by 50% or more compared to those before drug administration, while non-responders were those whose total HAMD scores had decreased by less than 50%. Here we showed that serum BDNF levels were not significantly different at any point between the two groups. The responder group, but not the non-responder group, showed statistically significant changes in serum BDNF 0 and serum BDNF 8. The results suggest that the changes of serum BDNF might differ between the two groups. The measurement of serum BDNF has the potential to be a useful predictor of pharmacotherapy in patients with first-episode, drug-naïve MD.
Collapse
|
48
|
Guo W, Yao X, Cui R, Yang W, Wang L. Mechanisms of paeoniaceae action as an antidepressant. Front Pharmacol 2023; 13:934199. [PMID: 36844911 PMCID: PMC9944447 DOI: 10.3389/fphar.2022.934199] [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: 05/02/2022] [Accepted: 08/05/2022] [Indexed: 02/10/2023] Open
Abstract
Paeoniflorin (PF) has been widely used for the treatment of depression in mice models, some Chinese herbal compound containing PF on treating depression, such as Xiaoyao San, Chaihu-Shugan-San, Danggui Shaoyao San etc. Many experiments are also verifying whether PF in these powders can be used as an effective component in the treatment of depression. Therefore, in this review the antidepressant effect of PF and its mechanism of action are outlined with particular focus on the following aspects: increasing the levels of monoamine neurotransmitters, inhibiting the HPA axis, promoting neuroprotection, enhancing neurogenesis in the hippocampus, and elevating levels of brain-derived neurotrophic factor (BDNF). This review may be helpful for the application of PF in the treatment of depression.
Collapse
Affiliation(s)
- Wanxu Guo
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Xiaoxiao Yao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Wei Yang
- *Correspondence: Wei Yang, ; Lei Wang,
| | - Lei Wang
- *Correspondence: Wei Yang, ; Lei Wang,
| |
Collapse
|
49
|
Rossi GN, Hallak JEC, Baker G, Dursun SM, Dos Santos RG. The effects of ketamine and classic hallucinogens on neurotrophic and inflammatory markers in unipolar treatment-resistant depression: a systematic review of clinical trials. Eur Arch Psychiatry Clin Neurosci 2023; 273:129-155. [PMID: 35829812 DOI: 10.1007/s00406-022-01460-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/27/2022] [Indexed: 11/03/2022]
Abstract
Although results are still preliminary, ketamine and classical hallucinogens have shown promise in recent years as novel, fast-acting antidepressants, especially for the treatment of unipolar treatment-resistant depression (TRD). Depression also seems to be related to abnormal levels of peripheral inflammatory and neurotrophic biomarkers, which may one day help to diagnose of this disorder. In this context, this systematic review of clinical trials evaluated the current evidence that relates the antidepressant effects of ketamine and classical hallucinogens on TRD with changes in inflammatory and neurotrophic biomarkers. Twelve studies were found (n = 587), 2 with oral ayahuasca (1 mL/kg) and 10 with ketamine (mostly intravenous 0.5 mg/kg) administration. Results for all biomarkers assessed were contradictory and thus inconclusive. Randomized controlled trials with bigger samples and higher statistical power are warranted to clarify if peripheral biomarkers can confidently be used to indicate and measure ketamine's and classical hallucinogens' antidepressant effect. The PROSPERO ID for this study is CRD42021249089.
Collapse
Affiliation(s)
- Giordano Novak Rossi
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Jaime E C Hallak
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil.,National Institute for Translational Medicine (INCT-TM), CNPq, São Paulo, Brazil.,Department of Psychiatry (Neurochemical Research Unit) and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Glen Baker
- National Institute for Translational Medicine (INCT-TM), CNPq, São Paulo, Brazil.,Department of Psychiatry (Neurochemical Research Unit) and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Serdar M Dursun
- National Institute for Translational Medicine (INCT-TM), CNPq, São Paulo, Brazil.,Department of Psychiatry (Neurochemical Research Unit) and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Rafael G Dos Santos
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil. .,National Institute for Translational Medicine (INCT-TM), CNPq, São Paulo, Brazil. .,Departamento de Neurociências e Ciências do Comportamento, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Hospital das Clínicas, Terceiro Andar, Av. Bandeirantes, 3900, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
50
|
Rodriguez LA, Kim SH, Page SC, Nguyen CV, Pattie EA, Hallock HL, Valerino J, Maynard KR, Jaffe AE, Martinowich K. The basolateral amygdala to lateral septum circuit is critical for regulating social novelty in mice. Neuropsychopharmacology 2023; 48:529-539. [PMID: 36369482 PMCID: PMC9852457 DOI: 10.1038/s41386-022-01487-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 10/07/2022] [Accepted: 10/24/2022] [Indexed: 11/13/2022]
Abstract
The lateral septum (LS) is a basal forebrain GABAergic region that is implicated in social novelty. However, the neural circuits and cell signaling pathways that converge on the LS to mediate social behaviors aren't well understood. Multiple lines of evidence suggest that signaling of brain-derived neurotrophic factor (BDNF) through its receptor TrkB plays important roles in social behavior. BDNF is not locally produced in LS, but we demonstrate that nearly all LS GABAergic neurons express TrkB. Local TrkB knock-down in LS neurons decreased social novelty recognition and reduced recruitment of neural activity in LS neurons in response to social novelty. Since BDNF is not synthesized in LS, we investigated which inputs to LS could serve as potential BDNF sources for controlling social novelty recognition. We demonstrate that selectively ablating inputs to LS from the basolateral amygdala (BLA), but not from ventral CA1 (vCA1), impairs social novelty recognition. Moreover, depleting BDNF selectively in BLA-LS projection neurons phenocopied the decrease in social novelty recognition caused by either local LS TrkB knockdown or ablation of BLA-LS inputs. These data support the hypothesis that BLA-LS projection neurons serve as a critical source of BDNF for activating TrkB signaling in LS neurons to control social novelty recognition.
Collapse
Affiliation(s)
- Lionel A Rodriguez
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Sun-Hong Kim
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Stephanie C Page
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Claudia V Nguyen
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Elizabeth A Pattie
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Henry L Hallock
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Jessica Valerino
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Kristen R Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Andrew E Jaffe
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Department of Genetic Medicine, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, 21205, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Keri Martinowich
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- The Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, USA.
| |
Collapse
|