1
|
Han M, Zeng D, Tan W, Chen X, Bai S, Wu Q, Chen Y, Wei Z, Mei Y, Zeng Y. Brain region-specific roles of brain-derived neurotrophic factor in social stress-induced depressive-like behavior. Neural Regen Res 2025; 20:159-173. [PMID: 38767484 PMCID: PMC11246125 DOI: 10.4103/nrr.nrr-d-23-01419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 01/19/2024] [Indexed: 05/22/2024] Open
Abstract
Brain-derived neurotrophic factor is a key factor in stress adaptation and avoidance of a social stress behavioral response. Recent studies have shown that brain-derived neurotrophic factor expression in stressed mice is brain region-specific, particularly involving the corticolimbic system, including the ventral tegmental area, nucleus accumbens, prefrontal cortex, amygdala, and hippocampus. Determining how brain-derived neurotrophic factor participates in stress processing in different brain regions will deepen our understanding of social stress psychopathology. In this review, we discuss the expression and regulation of brain-derived neurotrophic factor in stress-sensitive brain regions closely related to the pathophysiology of depression. We focused on associated molecular pathways and neural circuits, with special attention to the brain-derived neurotrophic factor-tropomyosin receptor kinase B signaling pathway and the ventral tegmental area-nucleus accumbens dopamine circuit. We determined that stress-induced alterations in brain-derived neurotrophic factor levels are likely related to the nature, severity, and duration of stress, especially in the above-mentioned brain regions of the corticolimbic system. Therefore, BDNF might be a biological indicator regulating stress-related processes in various brain regions.
Collapse
Affiliation(s)
- Man Han
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Deyang Zeng
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Wei Tan
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Xingxing Chen
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Shuyuan Bai
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Qiong Wu
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Yushan Chen
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Zhen Wei
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Yufei Mei
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Yan Zeng
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei Province, China
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
2
|
Oga K, Fuchikami M, Kobayashi H, Miyagi T, Fujita S, Fujita S, Okada S, Morinobu S. Involvement of dysregulated hippocampal histone H3K9 methylation at the promoter of the BDNF gene in impaired memory extinction. Psychopharmacology (Berl) 2024; 241:2363-2374. [PMID: 38940908 PMCID: PMC11513706 DOI: 10.1007/s00213-024-06640-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 06/17/2024] [Indexed: 06/29/2024]
Abstract
RATIONALE Since the precise mechanisms of posttraumatic stress disorder (PTSD) remain unknown, effective treatment interventions have not yet been established. Impaired extinction of fear memory (EFM) is one of the core symptoms of PTSD and is associated with stress-induced epigenetic change in gene expression. OBJECTIVES In this study, we examined whether the involvement of histone H3 lysine 9 dimethylation (H3K9me2) in EFM is mediated through brain-derived neurotrophic factor (BDNF) expression in the hippocampus, and whether BIX01294, a selective G9a and GLP histone methyltransferase inhibitor, could be treatment for impaired EFM in an animal model of PTSD. METHODS The single prolonged stress (SPS) paradigm was used to model PTSD. We measured BDNF mRNA levels by RT-PCR, and H3K9me2 levels in the BDNF gene promoters by chromatin immunoprecipitation-qPCR. After undergoing contextual fear conditioning and hippocampal injection of BIX01294, male rats were subjected to extinction training and extinction testing and their freezing times and BDNF mRNA levels were measured. RESULTS Compared to sham rats, SPS rats showed decreased BDNF mRNA levels 2 h after extinction training, no significant changes in levels of global H3K9me2 prior to extinction training, and increased levels of H3K9me2 in BDNF gene promoter IV, but not in BDNF gene promoter I. Administration of BIX01294 ameliorated the decrease in BDNF mRNA levels 2 h after extinction training and subsequently alleviated impaired EFM in extinction tests in SPS rats. CONCLUSION We conclude that reduced hippocampal levels of BDNF mRNA due to increase in H3K9me2 levels may play a role in PTSD-associated EFM impairment, and BIX01294 could be a PTSD treatment option.
Collapse
Affiliation(s)
- Kenichi Oga
- Department of Psychiatry and Neuroscience, Division of Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-Ku, Kasumi 1-2-3, Hiroshima City, Hiroshima, Japan
| | - Manabu Fuchikami
- Department of Psychiatry and Neuroscience, Division of Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-Ku, Kasumi 1-2-3, Hiroshima City, Hiroshima, Japan.
| | - Hironori Kobayashi
- Department of Psychiatry and Neuroscience, Division of Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-Ku, Kasumi 1-2-3, Hiroshima City, Hiroshima, Japan
| | - Tatsuhiro Miyagi
- Department of Psychiatry and Neuroscience, Division of Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-Ku, Kasumi 1-2-3, Hiroshima City, Hiroshima, Japan
| | - Sho Fujita
- Department of Psychiatry and Neuroscience, Division of Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-Ku, Kasumi 1-2-3, Hiroshima City, Hiroshima, Japan
| | - Satoshi Fujita
- Department of Psychiatry and Neuroscience, Division of Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-Ku, Kasumi 1-2-3, Hiroshima City, Hiroshima, Japan
| | - Satoshi Okada
- Department of Psychiatry and Neuroscience, Division of Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-Ku, Kasumi 1-2-3, Hiroshima City, Hiroshima, Japan
| | - Shigeru Morinobu
- Department of Psychology, School of Faculty of Health and Wellness Sciences, Hiroshima International University, Kure, Japan
| |
Collapse
|
3
|
Yao W, Hu X, Wang X. Crossing epigenetic frontiers: the intersection of novel histone modifications and diseases. Signal Transduct Target Ther 2024; 9:232. [PMID: 39278916 PMCID: PMC11403012 DOI: 10.1038/s41392-024-01918-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/11/2024] [Accepted: 06/30/2024] [Indexed: 09/18/2024] Open
Abstract
Histone post-translational modifications (HPTMs), as one of the core mechanisms of epigenetic regulation, are garnering increasing attention due to their close association with the onset and progression of diseases and their potential as targeted therapeutic agents. Advances in high-throughput molecular tools and the abundance of bioinformatics data have led to the discovery of novel HPTMs which similarly affect gene expression, metabolism, and chromatin structure. Furthermore, a growing body of research has demonstrated that novel histone modifications also play crucial roles in the development and progression of various diseases, including various cancers, cardiovascular diseases, infectious diseases, psychiatric disorders, and reproductive system diseases. This review defines nine novel histone modifications: lactylation, citrullination, crotonylation, succinylation, SUMOylation, propionylation, butyrylation, 2-hydroxyisobutyrylation, and 2-hydroxybutyrylation. It comprehensively introduces the modification processes of these nine novel HPTMs, their roles in transcription, replication, DNA repair and recombination, metabolism, and chromatin structure, as well as their involvement in promoting the occurrence and development of various diseases and their clinical applications as therapeutic targets and potential biomarkers. Moreover, this review provides a detailed overview of novel HPTM inhibitors targeting various targets and their emerging strategies in the treatment of multiple diseases while offering insights into their future development prospects and challenges. Additionally, we briefly introduce novel epigenetic research techniques and their applications in the field of novel HPTM research.
Collapse
Affiliation(s)
- Weiyi Yao
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Xinting Hu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
- Taishan Scholars Program of Shandong Province, Jinan, Shandong, 250021, China.
| |
Collapse
|
4
|
De Simone S, Alfieri L, Bosco MA, Cantatore S, Carpinteri M, Cipolloni L, Neri M. The forensic aspects of suicide and neurotrophin factors: a research study. Front Pharmacol 2024; 15:1392832. [PMID: 39170712 PMCID: PMC11335659 DOI: 10.3389/fphar.2024.1392832] [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: 02/28/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
Introduction: Suicide represents a significant public health problem whose neurobiology is not yet fully understood. In many cases, suicidal behavior and psychiatric spectrum disorders are linked, in particular, to major depression. An emerging pathophysiological hypothesis underlines the role of neurotrophic factors, proteins involved in neurogenesis, in synaptic plasticity in response to stressors. Our research aims to evaluate the degree of expression of brain neurotrophic factor (BDNF) in brain areas involved in depressive disorder in suicidal subjects. Furthermore, we want to evaluate the expression of glial cell line-derived neurotrophic factor (GDNF) in suicidal subjects. Methods: We selected twenty confirmed cases of suicide among subjects with a clinical history of depressive pathology and possible psychopharmacological treatment, compared to ten controls of individuals who died of non-suicidal causes. For all selected cases and controls, immunohistochemical investigations were performed using a panel of antibodies against the BDNF and GDNF antigens on samples from the various brain areas. Results and discussion: The results show that BDNF was under-expressed in the cerebral parenchyma of subjects who died by suicide compared to controls, while there was an overexpression of GDNF in suicide victims, these data could be useful for a clinical application as potential markers for suicidal risk, to assess the severity of depression and development of specific pharmacological therapies for depression.
Collapse
Affiliation(s)
- Stefania De Simone
- Department of Clinical and Experimental Medicine, Section of Legal Medicine, University of Foggia, Foggia, Italy
| | - Letizia Alfieri
- Department of Medical Sciences, Section of Legal Medicine University of Ferrara, Ferrara, Italy
| | - Maria Antonella Bosco
- Department of Clinical and Experimental Medicine, Section of Legal Medicine, University of Foggia, Foggia, Italy
| | - Santina Cantatore
- Department of Clinical and Experimental Medicine, Section of Legal Medicine, University of Foggia, Foggia, Italy
| | - Michele Carpinteri
- Department of Biomedical, Metabolic and Neural Sciences, Institute of Legal Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Luigi Cipolloni
- Department of Clinical and Experimental Medicine, Section of Legal Medicine, University of Foggia, Foggia, Italy
| | - Margherita Neri
- Department of Medical Sciences, Section of Legal Medicine University of Ferrara, Ferrara, Italy
| |
Collapse
|
5
|
Abbasi N, Mirabzadeh Y, Khesali G, Ebrahimkhani Z, Karimi H, Vaseghi S. Chronic REM sleep deprivation leads to manic- and OCD-related behaviors, and decreases hippocampal BDNF expression in female rats. Psychopharmacology (Berl) 2024; 241:1345-1363. [PMID: 38430395 DOI: 10.1007/s00213-024-06566-0] [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: 06/20/2023] [Accepted: 02/22/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND Rapid-eye movement (REM) sleep deprivation (SD) can induce manic-like behaviors in rodents. On the other hand, lithium, as one of the oldest drugs used in neuropsychiatric disorders, is still one of the best drugs for the treatment and control of bipolar disorder. In this study, we aimed to investigate the role of chronic short-term REM SD in the induction of manic-like behaviors in female rats. METHODS The rats were exposed to REM SD for 14 days (6 hours/day). Lithium was intraperitoneally injected at the doses of 10, 50, and 100 mg/kg. RESULTS REM SD induced hyperactivity and OCD-like behavior, and decreased anxiety, depressive-like behavior, and pain subthreshold. REM SD also impaired passive avoidance memory and decreased hippocampal brain-derived neurotrophic factor (BDNF) expression level. Lithium at the doses of 50 and 100 mg/kg partly and completely abolished these effects, respectively. However, lithium (100 mg/kg) increased BDNF expression level in control and sham REM SD rats with no significant changes in behavior. CONCLUSIONS Chronic short-term REM SD may induce a mania-like model and lead to OCD-like behavior and irritability. In the present study, we demonstrated a putative rodent model of mania induced by chronic REM SD in female rats. We suggest that future studies should examine behavioral and mood changes following chronic REM SD in both sexes. Furthermore, the relationship between manic-like behaviors and chronic REM SD should be investigated.
Collapse
Affiliation(s)
- Nahal Abbasi
- Department of Health Psychology, Faculty of Medical Sciences, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Yasaman Mirabzadeh
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Golnaz Khesali
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Zahra Ebrahimkhani
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Hanie Karimi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Salar Vaseghi
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran.
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran.
| |
Collapse
|
6
|
Bortolozzi A, Fico G, Berk M, Solmi M, Fornaro M, Quevedo J, Zarate CA, Kessing LV, Vieta E, Carvalho AF. New Advances in the Pharmacology and Toxicology of Lithium: A Neurobiologically Oriented Overview. Pharmacol Rev 2024; 76:323-357. [PMID: 38697859 PMCID: PMC11068842 DOI: 10.1124/pharmrev.120.000007] [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: 07/06/2020] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 05/05/2024] Open
Abstract
Over the last six decades, lithium has been considered the gold standard treatment for the long-term management of bipolar disorder due to its efficacy in preventing both manic and depressive episodes as well as suicidal behaviors. Nevertheless, despite numerous observed effects on various cellular pathways and biologic systems, the precise mechanism through which lithium stabilizes mood remains elusive. Furthermore, there is recent support for the therapeutic potential of lithium in other brain diseases. This review offers a comprehensive examination of contemporary understanding and predominant theories concerning the diverse mechanisms underlying lithium's effects. These findings are based on investigations utilizing cellular and animal models of neurodegenerative and psychiatric disorders. Recent studies have provided additional support for the significance of glycogen synthase kinase-3 (GSK3) inhibition as a crucial mechanism. Furthermore, research has shed more light on the interconnections between GSK3-mediated neuroprotective, antioxidant, and neuroplasticity processes. Moreover, recent advancements in animal and human models have provided valuable insights into how lithium-induced modifications at the homeostatic synaptic plasticity level may play a pivotal role in its clinical effectiveness. We focused on findings from translational studies suggesting that lithium may interface with microRNA expression. Finally, we are exploring the repurposing potential of lithium beyond bipolar disorder. These recent findings on the therapeutic mechanisms of lithium have provided important clues toward developing predictive models of response to lithium treatment and identifying new biologic targets. SIGNIFICANCE STATEMENT: Lithium is the drug of choice for the treatment of bipolar disorder, but its mechanism of action in stabilizing mood remains elusive. This review presents the latest evidence on lithium's various mechanisms of action. Recent evidence has strengthened glycogen synthase kinase-3 (GSK3) inhibition, changes at the level of homeostatic synaptic plasticity, and regulation of microRNA expression as key mechanisms, providing an intriguing perspective that may help bridge the mechanistic gap between molecular functions and its clinical efficacy as a mood stabilizer.
Collapse
Affiliation(s)
- Analia Bortolozzi
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Giovanna Fico
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Michael Berk
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Marco Solmi
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Michele Fornaro
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Joao Quevedo
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Carlos A Zarate
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Lars V Kessing
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Eduard Vieta
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Andre F Carvalho
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| |
Collapse
|
7
|
Bommaraju S, Dhokne MD, Arun EV, Srinivasan K, Sharma SS, Datusalia AK. An insight into crosstalk among multiple signalling pathways contributing to the pathophysiology of PTSD and depressive disorders. Prog Neuropsychopharmacol Biol Psychiatry 2024; 131:110943. [PMID: 38228244 DOI: 10.1016/j.pnpbp.2024.110943] [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: 08/05/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/18/2024]
Abstract
Post-traumatic stress disorder (PTSD) and depressive disorders represent two significant mental health challenges with substantial global prevalence. These are debilitating conditions characterized by persistent, often comorbid, symptoms that severely impact an individual's quality of life. Both PTSD and depressive disorders are often precipitated by exposure to traumatic events or chronic stress. The profound impact of PTSD and depressive disorders on individuals and society necessitates a comprehensive exploration of their shared and distinct pathophysiological features. Although the activation of the stress system is essential for maintaining homeostasis, the ability to recover from it after diminishing the threat stimulus is also equally important. However, little is known about the main reasons for individuals' differential susceptibility to external stressful stimuli. The solution to this question can be found by delving into the interplay of stress with the cognitive and emotional processing of traumatic incidents at the molecular level. Evidence suggests that dysregulation in these signalling cascades may contribute to the persistence and severity of PTSD and depressive symptoms. The treatment strategies available for this disorder are antidepressants, which have shown good efficiency in normalizing symptom severity; however, their efficacy is limited in most individuals. This calls for the exploration and development of innovative medications to address the treatment of PTSD. This review delves into the intricate crosstalk among multiple signalling pathways implicated in the development and manifestation of these mental health conditions. By unravelling the complexities of crosstalk among multiple signalling pathways, this review aims to contribute to the broader knowledge base, providing insights that could inform the development of targeted interventions for individuals grappling with the challenges of PTSD and depressive disorders.
Collapse
Affiliation(s)
- Sumadhura Bommaraju
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Uttar Pradesh (UP) 226002, India
| | - Mrunali D Dhokne
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Uttar Pradesh (UP) 226002, India
| | - E V Arun
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Uttar Pradesh (UP) 226002, India
| | - Krishnamoorthy Srinivasan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), SAS Nagar, Punjab 160062, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), SAS Nagar, Punjab 160062, India
| | - Ashok Kumar Datusalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Uttar Pradesh (UP) 226002, India; Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Raebareli, Uttar Pradesh (UP) 226002, India.
| |
Collapse
|
8
|
Ribeiro-Davis A, Al Saeedy DY, Jahr FM, Hawkins E, McClay JL, Deshpande LS. Ketamine Produces Antidepressant Effects by Inhibiting Histone Deacetylases and Upregulating Hippocampal Brain-Derived Neurotrophic Factor Levels in a Diisopropyl Fluorophosphate-Based Rat Model of Gulf War Illness. J Pharmacol Exp Ther 2024; 388:647-654. [PMID: 37863487 PMCID: PMC10801753 DOI: 10.1124/jpet.123.001824] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 09/14/2023] [Accepted: 09/14/2023] [Indexed: 10/22/2023] Open
Abstract
Approximately one-third of Gulf War veterans suffer from Gulf War Illness (GWI), which encompasses mood disorders and depressive symptoms. Deployment-related exposure to organophosphate compounds has been associated with GWI development. Epigenetic modifications have been reported in GWI veterans. We previously showed that epigenetic histone dysregulations were associated with decreased brain-derived neurotrophic factor (BDNF) expression in a GWI rat model. GWI has no effective therapies. Ketamine (KET) has recently been approved by the Food and Drug Administration for therapy-resistant depression. Interestingly, BDNF upregulation underlies KET's antidepressant effect in GWI-related depression. Here, we investigated whether KET's effect on histone mechanisms signals BDNF upregulations in GWI. Male Sprague-Dawley rats were injected once daily with diisopropyl fluorophosphate (DFP; 0.5 mg/kg, s.c., 5 days). At 6 months following DFP exposure, KET (10 mg/kg, i.p.) was injected, and brains were dissected 24 hours later. Western blotting was used for protein expression, and epigenetic studies used chromatin immunoprecipitation methods. Dil staining was conducted for assessing dendritic spines. Our results indicated that an antidepressant dose of KET inhibited the upregulation of histone deacetylase (HDAC) enzymes in DFP rats. Furthermore, KET restored acetylated histone occupancy at the Bdnf promoter IV and induced BDNF protein expression in DFP rats. Finally, KET treatment also increased the spine density and altered the spine diversity with increased T-type and decreased S-type spines in DFP rats. Given these findings, we propose that KET's actions involve the inhibition of HDAC expression, upregulation of BDNF, and dendritic modifications that together ameliorates the pathologic synaptic plasticity and exerts an antidepressant effect in DFP rats. SIGNIFICANCE STATEMENT: This study offers evidence supporting the involvement of epigenetic histone pathways in the antidepressant effects of ketamine (KET) in a rat model of Gulf War Illness (GWI)-like depression. This effect is achieved through the modulation of histone acetylation at the Bdnf promoter, resulting in elevated brain-derived neurotrophic factor expression and subsequent dendritic remodeling in the hippocampus. These findings underscore the rationale for considering KET as a potential candidate for clinical trials aimed at managing GWI-related depression.
Collapse
Affiliation(s)
- Ana Ribeiro-Davis
- Departments of Neurology (A.R.-D., E.H., L.S.D.), Pharmacology and Toxicology (L.S.D.), School of Medicine, Virginia Commonwealth University, Richmond, Virginia and Department of Pharmacotherapy and Outcome Sciences (D.Y.A.S., F.M.J., J.L.M.), School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Dalia Y Al Saeedy
- Departments of Neurology (A.R.-D., E.H., L.S.D.), Pharmacology and Toxicology (L.S.D.), School of Medicine, Virginia Commonwealth University, Richmond, Virginia and Department of Pharmacotherapy and Outcome Sciences (D.Y.A.S., F.M.J., J.L.M.), School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Fay M Jahr
- Departments of Neurology (A.R.-D., E.H., L.S.D.), Pharmacology and Toxicology (L.S.D.), School of Medicine, Virginia Commonwealth University, Richmond, Virginia and Department of Pharmacotherapy and Outcome Sciences (D.Y.A.S., F.M.J., J.L.M.), School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Elisa Hawkins
- Departments of Neurology (A.R.-D., E.H., L.S.D.), Pharmacology and Toxicology (L.S.D.), School of Medicine, Virginia Commonwealth University, Richmond, Virginia and Department of Pharmacotherapy and Outcome Sciences (D.Y.A.S., F.M.J., J.L.M.), School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Joseph L McClay
- Departments of Neurology (A.R.-D., E.H., L.S.D.), Pharmacology and Toxicology (L.S.D.), School of Medicine, Virginia Commonwealth University, Richmond, Virginia and Department of Pharmacotherapy and Outcome Sciences (D.Y.A.S., F.M.J., J.L.M.), School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Laxmikant S Deshpande
- Departments of Neurology (A.R.-D., E.H., L.S.D.), Pharmacology and Toxicology (L.S.D.), School of Medicine, Virginia Commonwealth University, Richmond, Virginia and Department of Pharmacotherapy and Outcome Sciences (D.Y.A.S., F.M.J., J.L.M.), School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
9
|
Dell'Osso L, Nardi B, Massoni L, Gravina D, Benedetti F, Cremone IM, Carpita B. Neuroprotective Properties of Antiepileptics: What are the Implications for Psychiatric Disorders? Curr Med Chem 2024; 31:3447-3472. [PMID: 37226791 DOI: 10.2174/0929867330666230523155728] [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: 12/31/2022] [Revised: 03/29/2023] [Accepted: 04/14/2023] [Indexed: 05/26/2023]
Abstract
Since the discovery of the first antiepileptic compound, increasing attention has been paid to antiepileptic drugs (AEDs), and recently, with the understanding of the molecular mechanism underlying cells death, a new interest has revolved around a potential neuroprotective effect of AEDs. While many neurobiological studies in this field have focused on the protection of neurons, growing data are reporting how exposure to AEDs can also affect glial cells and the plastic response underlying recovery; however, demonstrating the neuroprotective abilities of AEDs remains a changeling task. The present work aims to summarize and review the literature available on the neuroprotective properties of the most commonly used AEDs. Results highlighted how further studies should investigate the link between AEDs and neuroprotective properties; while many studies are available on valproate, results for other AEDs are very limited and the majority of the research has been carried out on animal models. Moreover, a better understanding of the biological basis underlying neuro-regenerative defects may pave the way for the investigation of further therapeutic targets and eventually lead to an improvement in the actual treatment strategies.
Collapse
Affiliation(s)
- Liliana Dell'Osso
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Benedetta Nardi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Leonardo Massoni
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Davide Gravina
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Francesca Benedetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Ivan Mirko Cremone
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Barbara Carpita
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| |
Collapse
|
10
|
Jastrzębski MK, Wójcik P, Stępnicki P, Kaczor AA. Effects of small molecules on neurogenesis: Neuronal proliferation and differentiation. Acta Pharm Sin B 2024; 14:20-37. [PMID: 38239239 PMCID: PMC10793103 DOI: 10.1016/j.apsb.2023.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 09/06/2023] [Accepted: 09/13/2023] [Indexed: 01/22/2024] Open
Abstract
Neurons are believed to be non-proliferating cells. However, neuronal stem cells are still present in certain areas of the adult brain, although their proliferation diminishes with age. Just as with other cells, their proliferation and differentiation are modulated by various mechanisms. These mechanisms are foundational to the strategies developed to induce neuronal proliferation and differentiation, with potential therapeutic applications for neurodegenerative diseases. The most common among these diseases are Parkinson's disease and Alzheimer's disease, associated with the formation of β -amyloid (Aβ ) aggregates which cause a reduction in the number of neurons. Compounds such as LiCl, 4-aminothiazoles, Pregnenolone, ACEA, harmine, D2AAK1, methyl 3,4-dihydroxybenzoate, and shikonin may induce neuronal proliferation/differentiation through the activation of pathways: MAPK ERK, PI3K/AKT, NFκ B, Wnt, BDNF, and NPAS3. Moreover, combinations of these compounds can potentially transform somatic cells into neurons. This transformation process involves the activation of neuron-specific transcription factors such as NEUROD1, NGN2, ASCL1, and SOX2, which subsequently leads to the transcription of downstream genes, culminating in the transformation of somatic cells into neurons. Neurodegenerative diseases are not the only conditions where inducing neuronal proliferation could be beneficial. Consequently, the impact of pro-proliferative compounds on neurons has also been researched in mouse models of Alzheimer's disease.
Collapse
Affiliation(s)
- Michał K. Jastrzębski
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Laboratory, Medical University of Lublin, Faculty of Pharmacy, Lublin PL-20093, Poland
| | - Piotr Wójcik
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Laboratory, Medical University of Lublin, Faculty of Pharmacy, Lublin PL-20093, Poland
| | - Piotr Stępnicki
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Laboratory, Medical University of Lublin, Faculty of Pharmacy, Lublin PL-20093, Poland
| | - Agnieszka A. Kaczor
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Laboratory, Medical University of Lublin, Faculty of Pharmacy, Lublin PL-20093, Poland
- School of Pharmacy, University of Eastern Finland, Kuopio FI-70211, Finland
| |
Collapse
|
11
|
Liang Y, Kang X, Zhang H, Xu H, Wu X. Knockdown and inhibition of hippocampal GPR17 attenuates lipopolysaccharide-induced cognitive impairment in mice. J Neuroinflammation 2023; 20:271. [PMID: 37990234 PMCID: PMC10662506 DOI: 10.1186/s12974-023-02958-9] [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: 08/11/2023] [Accepted: 11/13/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND Previously we reported that inhibition of GPR17 prevents amyloid β 1-42 (Aβ1-42)-induced cognitive impairment in mice. However, the role of GPR17 on cognition is still largely unknown. METHODS Herein, we used a mouse model of cognitive impairment induced by lipopolysaccharide (LPS) to further investigate the role of GPR17 in cognition and its potential mechanism. The mice were pretreated with GPR17 shRNA lentivirus and cangrelor by microinjection into the dentate gyrus (DG) region of the hippocampus. After 21 days, LPS (0.25 mg/kg, i.p.) was administered for 7 days. Animal behavioral tests as well as pathological and biochemical assays were performed to evaluate the cognitive function in mice. RESULTS LPS exposure resulted in a significant increase in GPR17 expression at both protein and mRNA levels in the hippocampus. Gene reduction and pharmacological blockade of GPR17 improved cognitive impairment in both the Morris water maze and novel object recognition tests. Knockdown and inhibition of GPR17 inhibited Aβ production, decreased the expression of NF-κB p65, increased CREB phosphorylation and elevated BDNF expression, suppressed the accumulation of pro-inflammatory cytokines, inhibited Glial cells (microglia and astrocytes) activation, and increased Bcl-2, PSD-95, and SYN expression, reduced Bax expression as well as decreased caspase-3 activity and TUNEL-positive cells in the hippocampus of LPS-treated mice. Notably, knockdown and inhibition of GPR17 not only provided protective effects against cholinergic dysfunction but also facilitated the regulation of oxidative stress. In addition, cangrelor pretreatment can effectively inhibit the expression of inflammatory cytokines by suppressing NF-κB/CREB/BDNF signaling in BV-2 cells stimulated by LPS. However, activation of hippocampal GPR17 with MDL-29951 induced cognitive impairment in normal mice. CONCLUSIONS These observations indicate that GPR17 may possess a neuroprotective effect against LPS-induced cognition deficits, and neuroinflammation by modulation of NF-κB/CREB/BDNF signaling in mice, indicating that GPR17 may be a promising new target for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Yusheng Liang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Xu Kang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Haiwang Zhang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Heng Xu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Xian Wu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China.
| |
Collapse
|
12
|
Mrozek W, Socha J, Sidorowicz K, Skrok A, Syrytczyk A, Piątkowska-Chmiel I, Herbet M. Pathogenesis and treatment of depression: Role of diet in prevention and therapy. Nutrition 2023; 115:112143. [PMID: 37562078 PMCID: PMC10299949 DOI: 10.1016/j.nut.2023.112143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 06/20/2023] [Accepted: 06/23/2023] [Indexed: 08/12/2023]
Abstract
In recent years, there has been a significant increase in depression, which is related to, among other things, the COVID-19 pandemic. Depression can be fatal if not treated or if treated inappropriately. Depression is the leading cause of suicide attempts. The disease is multifactorial, and pharmacotherapy often fails to bring satisfactory results. Therefore, increasingly more importance is attached to the natural healing substances and nutrients in food, which can significantly affect the therapy process and prevention of depressive disorders. A proper diet is vital to preventing depression and can be a valuable addition to psychological and pharmacologic treatment. An inadequate diet may reduce the effectiveness of antidepressants or increase their side effects, leading to life-threatening symptoms. This study aimed to review the literature on the pathogenesis of the development and treatment of depression, with particular emphasis on dietary supplements and the role of nutrition in the prevention and treatment of depressive disorders.
Collapse
Affiliation(s)
- Weronika Mrozek
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Justyna Socha
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Klara Sidorowicz
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Aleksandra Skrok
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Aleksandra Syrytczyk
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | | | - Mariola Herbet
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland.
| |
Collapse
|
13
|
Ornell F, Scherer JN, Schuch JB, Sordi AO, Halpern SC, Rebelatto FP, Bristot G, Kapczinski F, Roglio VS, Pechansky F, Kessler FHP, von Diemen L. Serum BDNF levels increase during early drug withdrawal in alcohol and crack cocaine addiction. Alcohol 2023; 111:1-7. [PMID: 37037287 DOI: 10.1016/j.alcohol.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/02/2023] [Accepted: 04/05/2023] [Indexed: 04/12/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) is involved in several drug-induced brain neuroadaptations. The impact of withdrawal from substances that have different neurological mechanisms on BDNF levels is unclear. Our goal was to compare serum BDNF levels in inpatients with alcohol or crack cocaine use disorders during the early withdrawal period, and to evaluate the association with substance-related outcomes. We performed a follow-up study with 101 men under detoxification treatment (drug preference: alcohol [n = 37] and crack cocaine [n = 64]). Blood samples were collected on the 1st and 15th days of hospitalization to measure serum BDNF levels. Serum BDNF levels increased during the early stage of withdrawal (28.2 ± 10.0 vs. 32.6 ± 13.3, p < 0.001), similarly in individuals with alcohol and crack cocaine use. In the alcohol group, BDNF levels on the 15th day of hospitalization were negatively correlated with age (r = -0.394, p = 0.023). Delta BDNF levels were also negatively correlated with BDNF on the 1st day of hospitalization (p = 0.011). No significant correlation was found regarding substance-related outcomes. This is the first study to compare BDNF levels in alcohol and crack cocaine users undergoing similar treatment conditions. These findings could be related to clinical improvement after abstinence or even to drug withdrawal itself, decreasing neuronal injury. Furthermore, age may be a crucial factor, hindering the recovery of neuroplasticity in alcohol users.
Collapse
Affiliation(s)
- Felipe Ornell
- Center for Drug and Alcohol Research, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Psychiatry and Behavioral Sciences, Department of Psychiatry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Juliana N Scherer
- Center for Drug and Alcohol Research, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Jaqueline B Schuch
- Center for Drug and Alcohol Research, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Psychiatry and Behavioral Sciences, Department of Psychiatry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Anne O Sordi
- Center for Drug and Alcohol Research, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Silvia C Halpern
- Center for Drug and Alcohol Research, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernando P Rebelatto
- Center for Drug and Alcohol Research, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Psychiatry and Behavioral Sciences, Department of Psychiatry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Giovana Bristot
- Bipolar Disorder Program, Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil; Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Flavio Kapczinski
- Graduate Program in Psychiatry and Behavioral Sciences, Department of Psychiatry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Bipolar Disorder Program, Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil; Department of Psychiatry and Behavioural Neurosciences, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Vinicius S Roglio
- Center for Drug and Alcohol Research, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Psychiatry and Behavioral Sciences, Department of Psychiatry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Flavio Pechansky
- Center for Drug and Alcohol Research, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Psychiatry and Behavioral Sciences, Department of Psychiatry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Felix H P Kessler
- Center for Drug and Alcohol Research, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Psychiatry and Behavioral Sciences, Department of Psychiatry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Lisia von Diemen
- Center for Drug and Alcohol Research, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Psychiatry and Behavioral Sciences, Department of Psychiatry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
14
|
Arora S, Santiago JA, Bernstein M, Potashkin JA. Diet and lifestyle impact the development and progression of Alzheimer's dementia. Front Nutr 2023; 10:1213223. [PMID: 37457976 PMCID: PMC10344607 DOI: 10.3389/fnut.2023.1213223] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/08/2023] [Indexed: 07/18/2023] Open
Abstract
Dementia is a growing public health concern, with an estimated prevalence of 57 million adults worldwide. Alzheimer's disease (AD) accounts for 60-80% of the cases. Clinical trials testing potential drugs and neuroprotective agents have proven futile, and currently approved drugs only provide symptomatic benefits. Emerging epidemiological and clinical studies suggest that lifestyle changes, including diet and physical activity, offer an alternative therapeutic route for slowing and preventing cognitive decline and dementia. Age is the single most common risk factor for dementia, and it is associated with slowing cellular bioenergetics and metabolic processes. Therefore, a nutrient-rich diet is critical for optimal brain health. Furthermore, type 2 diabetes (T2D) is a risk factor for AD, and diets that reduce the risk of T2D may confer neuroprotection. Foods predominant in Mediterranean, MIND, and DASH diets, including fruits, leafy green vegetables, fish, nuts, and olive oil, may prevent or slow cognitive decline. The mechanisms by which these nutrients promote brain health, however, are not yet completely understood. Other dietary approaches and eating regimes, including ketogenic and intermittent fasting, are also emerging as beneficial for brain health. This review summarizes the pathophysiology, associated risk factors, and the potential neuroprotective pathways activated by several diets and eating regimes that have shown promising results in promoting brain health and preventing dementia.
Collapse
Affiliation(s)
- Sarah Arora
- Center for Neurodegenerative Diseases and Therapeutics, Cellular and Molecular Pharmacology Discipline, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | | | - Melissa Bernstein
- Department of Nutrition, College of Health Professions, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Judith A. Potashkin
- Center for Neurodegenerative Diseases and Therapeutics, Cellular and Molecular Pharmacology Discipline, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| |
Collapse
|
15
|
Gladkova MG, Leidmaa E, Anderzhanova EA. Epidrugs in the Therapy of Central Nervous System Disorders: A Way to Drive on? Cells 2023; 12:1464. [PMID: 37296584 PMCID: PMC10253154 DOI: 10.3390/cells12111464] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 05/01/2023] [Accepted: 05/16/2023] [Indexed: 06/12/2023] Open
Abstract
The polygenic nature of neurological and psychiatric syndromes and the significant impact of environmental factors on the underlying developmental, homeostatic, and neuroplastic mechanisms suggest that an efficient therapy for these disorders should be a complex one. Pharmacological interventions with drugs selectively influencing the epigenetic landscape (epidrugs) allow one to hit multiple targets, therefore, assumably addressing a wide spectrum of genetic and environmental mechanisms of central nervous system (CNS) disorders. The aim of this review is to understand what fundamental pathological mechanisms would be optimal to target with epidrugs in the treatment of neurological or psychiatric complications. To date, the use of histone deacetylases and DNA methyltransferase inhibitors (HDACis and DNMTis) in the clinic is focused on the treatment of neoplasms (mainly of a glial origin) and is based on the cytostatic and cytotoxic actions of these compounds. Preclinical data show that besides this activity, inhibitors of histone deacetylases, DNA methyltransferases, bromodomains, and ten-eleven translocation (TET) proteins impact the expression of neuroimmune inflammation mediators (cytokines and pro-apoptotic factors), neurotrophins (brain-derived neurotropic factor (BDNF) and nerve growth factor (NGF)), ion channels, ionotropic receptors, as well as pathoproteins (β-amyloid, tau protein, and α-synuclein). Based on this profile of activities, epidrugs may be favorable as a treatment for neurodegenerative diseases. For the treatment of neurodevelopmental disorders, drug addiction, as well as anxiety disorders, depression, schizophrenia, and epilepsy, contemporary epidrugs still require further development concerning a tuning of pharmacological effects, reduction in toxicity, and development of efficient treatment protocols. A promising strategy to further clarify the potential targets of epidrugs as therapeutic means to cure neurological and psychiatric syndromes is the profiling of the epigenetic mechanisms, which have evolved upon actions of complex physiological lifestyle factors, such as diet and physical exercise, and which are effective in the management of neurodegenerative diseases and dementia.
Collapse
Affiliation(s)
- Marina G. Gladkova
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Este Leidmaa
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, 53127 Bonn, Germany
- Institute of Biomedicine and Translational Medicine, Department of Physiology, University of Tartu, 50411 Tartu, Estonia
| | | |
Collapse
|
16
|
Inhibition of Microglial GSK3β Activity Is Common to Different Kinds of Antidepressants: A Proposal for an In Vitro Screen to Detect Novel Antidepressant Principles. Biomedicines 2023; 11:biomedicines11030806. [PMID: 36979785 PMCID: PMC10045655 DOI: 10.3390/biomedicines11030806] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/17/2023] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
Depression is a major public health concern. Unfortunately, the present antidepressants often are insufficiently effective, whilst the discovery of more effective antidepressants has been extremely sluggish. The objective of this review was to combine the literature on depression with the pharmacology of antidepressant compounds, in order to formulate a conceivable pathophysiological process, allowing proposals how to accelerate the discovery process. Risk factors for depression initiate an infection-like inflammation in the brain that involves activation microglial Toll-like receptors and glycogen synthase kinase-3β (GSK3β). GSK3β activity alters the balance between two competing transcription factors, the pro-inflammatory/pro-oxidative transcription factor NFκB and the neuroprotective, anti-inflammatory and anti-oxidative transcription factor NRF2. The antidepressant activity of tricyclic antidepressants is assumed to involve activation of GS-coupled microglial receptors, raising intracellular cAMP levels and activation of protein kinase A (PKA). PKA and similar kinases inhibit the enzyme activity of GSK3β. Experimental antidepressant principles, including cannabinoid receptor-2 activation, opioid μ receptor agonists, 5HT2 agonists, valproate, ketamine and electrical stimulation of the Vagus nerve, all activate microglial pathways that result in GSK3β-inhibition. An in vitro screen for NRF2-activation in microglial cells with TLR-activated GSK3β activity, might therefore lead to the detection of totally novel antidepressant principles with, hopefully, an improved therapeutic efficacy.
Collapse
|
17
|
Puglisi-Allegra S, Lazzeri G, Busceti CL, Giorgi FS, Biagioni F, Fornai F. Lithium engages autophagy for neuroprotection and neuroplasticity: translational evidence for therapy. Neurosci Biobehav Rev 2023; 148:105148. [PMID: 36996994 DOI: 10.1016/j.neubiorev.2023.105148] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/23/2023] [Accepted: 03/26/2023] [Indexed: 03/30/2023]
Abstract
Here an overview is provided on therapeutic/neuroprotective effects of Lithium (Li+) in neurodegenerative and psychiatric disorders focusing on the conspicuous action of Li+ through autophagy. The effects on the autophagy machinery remain the key molecular mechanisms to explain the protective effects of Li+ for neurodegenerative diseases, offering potential therapeutic strategies for the treatment of neuropsychiatric disorders and emphasizes a crossroad linking autophagy, neurodegenerative disorders, and mood stabilization. Sensitization by psychostimulants points to several mechanisms involved in psychopathology, most also crucial in neurodegenerative disorders. Evidence shows the involvement of autophagy and metabotropic Glutamate receptors-5 (mGluR5) in neurodegeneration due to methamphetamine neurotoxicity as well as in neuroprotection, both in vitro and in vivo models. More recently, Li+ was shown to modulate autophagy through its action on mGluR5, thus pointing to an additional way of autophagy engagement by Li+ and to a substantial role of mGluR5 in neuroprotection related to neural e neuropsychiatry diseases. We propose Li+ engagement of autophagy through the canonical mechanisms of autophagy machinery and through the intermediary of mGluR5.
Collapse
|
18
|
Thela L, Decloedt E, Zetterberg H, Gisslén M, Lesosky M, Gleich M, Koutsilieri E, Scheller C, Hye A, Joska J. Blood and cerebrospinal fluid biomarker changes in patients with HIV-associated neurocognitive impairment treated with lithium: analysis from a randomised placebo-controlled trial. J Neurovirol 2023; 29:156-166. [PMID: 36790601 DOI: 10.1007/s13365-023-01116-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 12/15/2022] [Accepted: 01/21/2023] [Indexed: 02/16/2023]
Abstract
HIV-associated neurocognitive disorders (HAND) persist in the era of antiretroviral therapy (ART). Thus, ART does not completely halt or reverse the pathological processes behind HAND. Adjuvant mitigating treatments are, therefore, prudent. Lithium treatment is known to promote neuronal brain-derived neurotrophic factors (BDNF). Lithium is also an inhibitor of glycogen synthase kinase-3 beta (GSK-3-β). We analyzed biomarkers obtained from participants in a randomized placebo-controlled trial of lithium in ART-treated individuals with moderate or severe HAND. We assayed markers at baseline and 24 weeks across several pathways hypothesized to be affected by HIV, inflammation, or degeneration. Investigated biomarkers included dopamine, BDNF, neurofilament light chain, and CD8 + lymphocyte activation (CD38 + HLADR +). Alzheimer's Disease (AD) biomarkers included soluble amyloid precursor protein alpha and beta (sAPPα/β), Aβ38, 40, 42, and ten other biomarkers validated as predictors of mild cognitive impairment and progression in previous studies. These include apolipoprotein C3, pre-albumin, α1-acid glycoprotein, α1-antitrypsin, PEDF, CC4, ICAM-1, RANTES, clusterin, and cystatin c. We recruited 61 participants (placebo = 31; lithium = 30). The age baseline mean was 40 (± 8.35) years and the median CD4 + T-cell count was 498 (IQR: 389-651) cells/μL. Biomarker concentrations between groups did not differ at baseline. However, both groups' blood dopamine levels decreased significantly after 24 weeks (adj. p < 002). No other marker was significantly different between groups, and we concluded that lithium did not confer neuroprotection following 24 weeks of treatment. However, the study was limited in duration and sample size.
Collapse
Affiliation(s)
- Lindokuhle Thela
- HIV Mental Health Research Unit, Department of Psychiatry and Mental Health, Neuroscience Institute, University of Cape Town, E Floor, Neuroscience Centre, Anzio Road, Groote Schuur Hospital, Observatory, 7925, Cape Town, South Africa.
| | - Eric Decloedt
- Division of Clinical Pharmacology, Department of Medicine, Stellenbosch University, Cape Town, South Africa
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK.,UK Dementia Research Institute at UCL, London, UK.,Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Magnus Gisslén
- Department of Infectious Disease, Institute of Biomedicine, the Sahlngreska Academy at the University of Gothenburg, Gothenburg, Sweden.,Department of Infectious Disease, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Maia Lesosky
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Melanie Gleich
- Institute of Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Eleni Koutsilieri
- Institute of Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Carsten Scheller
- Institute of Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Abdul Hye
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, and NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | - John Joska
- HIV Mental Health Research Unit, Department of Psychiatry and Mental Health, Neuroscience Institute, University of Cape Town, E Floor, Neuroscience Centre, Anzio Road, Groote Schuur Hospital, Observatory, 7925, Cape Town, South Africa
| |
Collapse
|
19
|
Lee K, Mills Z, Cheung P, Cheyne JE, Montgomery JM. The Role of Zinc and NMDA Receptors in Autism Spectrum Disorders. Pharmaceuticals (Basel) 2022; 16:ph16010001. [PMID: 36678498 PMCID: PMC9866730 DOI: 10.3390/ph16010001] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
NMDA-type glutamate receptors are critical for synaptic plasticity in the central nervous system. Their unique properties and age-dependent arrangement of subunit types underpin their role as a coincidence detector of pre- and postsynaptic activity during brain development and maturation. NMDAR function is highly modulated by zinc, which is co-released with glutamate and concentrates in postsynaptic spines. Both NMDARs and zinc have been strongly linked to autism spectrum disorders (ASDs), suggesting that NMDARs are an important player in the beneficial effects observed with zinc in both animal models and children with ASDs. Significant evidence is emerging that these beneficial effects occur via zinc-dependent regulation of SHANK proteins, which form the backbone of the postsynaptic density. For example, dietary zinc supplementation enhances SHANK2 or SHANK3 synaptic recruitment and rescues NMDAR deficits and hypofunction in Shank3ex13-16-/- and Tbr1+/- ASD mice. Across multiple studies, synaptic changes occur in parallel with a reversal of ASD-associated behaviours, highlighting the zinc-dependent regulation of NMDARs and glutamatergic synapses as therapeutic targets for severe forms of ASDs, either pre- or postnatally. The data from rodent models set a strong foundation for future translational studies in human cells and people affected by ASDs.
Collapse
|
20
|
Chang HH, Hsueh YS, Cheng YW, Tseng HH. A Longitudinal Study of the Association between the LEPR Polymorphism and Treatment Response in Patients with Bipolar Disorder. Int J Mol Sci 2022; 23:ijms23179635. [PMID: 36077028 PMCID: PMC9455965 DOI: 10.3390/ijms23179635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/30/2022] Open
Abstract
Patients with bipolar disorder (BD) exhibit individual variability in the treatment outcome, and genetic background could contribute to BD itself and the treatment outcome. Leptin levels significantly change in BD patients treated with valproate (VPA), but whether LEPR polymorphisms are associated with treatment response is still unknown. This longitudinal study aimed to investigate the associations between LEPR polymorphisms and VPA treatment response in BD patients who were drug naïve at their first diagnosis of BD. The single-nucleotide polymorphisms (SNPs) of LEPR (rs1137101, rs1137100, rs8179183, and rs12145690) were assayed, and the LEPR polymorphism frequencies of alleles and genotypes were not significantly different between the controls (n = 77) and BD patients (n = 130). In addition, after the 12-week course of VPA treatment in BD patients, the LEPR polymorphisms showed significant effects on changes in disease severity. Moreover, considering the effect of the LEPR haplotype, the frequency of the CAGG haplotype in BD patients was higher than that in the controls (9.3 vs. 2.9%, p = 0.016), and the LEPR CAGG haplotype was associated with a better treatment response than the other haplotypes in BD patients receiving VPA treatment. Therefore, LEPR polymorphisms might serve as mediators involved in the therapeutic action of VPA treatment.
Collapse
Affiliation(s)
- Hui Hua Chang
- Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Department of Pharmacy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Department of Pharmacy, National Cheng Kung University Hospital, Dou-Liou Branch, Yunlin 640, Taiwan
- Correspondence: ; Tel.: +886-6-2353535 (ext. 5683)
| | - Yuan-Shuo Hsueh
- Department of Medical Science Industries, College of Health Sciences, Chang Jung Christian University, Tainan 711, Taiwan
| | - Yung Wen Cheng
- Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Huai-Hsuan Tseng
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| |
Collapse
|
21
|
Hung SY, Chung HY, Luo ST, Chu YT, Chen YH, MacDonald IJ, Chien SY, Kotha P, Yang LY, Hwang LL, Dun NJ, Chuang DM, Chen YH. Electroacupuncture improves TBI dysfunction by targeting HDAC overexpression and BDNF-associated Akt/GSK-3β signaling. Front Cell Neurosci 2022; 16:880267. [PMID: 36016833 PMCID: PMC9396337 DOI: 10.3389/fncel.2022.880267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 06/27/2022] [Indexed: 11/18/2022] Open
Abstract
Background Acupuncture or electroacupuncture (EA) appears to be a potential treatment in acute clinical traumatic brain injury (TBI); however, it remains uncertain whether acupuncture affects post-TBI histone deacetylase (HDAC) expression or impacts other biochemical/neurobiological events. Materials and methods We used behavioral testing, Western blot, and immunohistochemistry analysis to evaluate the cellular and molecular effects of EA at LI4 and LI11 in both weight drop-impact acceleration (WD)- and controlled cortical impact (CCI)-induced TBI models. Results Both WD- and CCI-induced TBI caused behavioral dysfunction, increased cortical levels of HDAC1 and HDAC3 isoforms, activated microglia and astrocytes, and decreased cortical levels of BDNF as well as its downstream mediators phosphorylated-Akt and phosphorylated-GSK-3β. Application of EA reversed motor, sensorimotor, and learning/memory deficits. EA also restored overexpression of HDAC1 and HDAC3, and recovered downregulation of BDNF-associated signaling in the cortex of TBI mice. Conclusion The results strongly suggest that acupuncture has multiple benefits against TBI-associated adverse behavioral and biochemical effects and that the underlying mechanisms are likely mediated by targeting HDAC overexpression and aberrant BDNF-associated Akt/GSK-3 signaling.
Collapse
Affiliation(s)
- Shih-Ya Hung
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
- Division of Colorectal Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Hsin-Yi Chung
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | - Sih-Ting Luo
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | - Yu-Ting Chu
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | - Yu-Hsin Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Iona J. MacDonald
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | - Szu-Yu Chien
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | - Peddanna Kotha
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | - Liang-Yo Yang
- Department of Physiology, School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
- Laboratory for Neural Repair, China Medical University Hospital, Taichung, Taiwan
| | - Ling-Ling Hwang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Nae J. Dun
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, United States
| | - De-Maw Chuang
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Yi-Hung Chen
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
- Department of Photonics and Communication Engineering, Asia University, Taichung, Taiwan
- *Correspondence: Yi-Hung Chen,
| |
Collapse
|
22
|
Nicoletti VG, Pajer K, Calcagno D, Pajenda G, Nógrádi A. The Role of Metals in the Neuroregenerative Action of BDNF, GDNF, NGF and Other Neurotrophic Factors. Biomolecules 2022; 12:biom12081015. [PMID: 35892326 PMCID: PMC9330237 DOI: 10.3390/biom12081015] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 12/14/2022] Open
Abstract
Mature neurotrophic factors and their propeptides play key roles ranging from the regulation of neuronal growth and differentiation to prominent participation in neuronal survival and recovery after injury. Their signaling pathways sculpture neuronal circuits during brain development and regulate adaptive neuroplasticity. In addition, neurotrophic factors provide trophic support for damaged neurons, giving them a greater capacity to survive and maintain their potential to regenerate their axons. Therefore, the modulation of these factors can be a valuable target for treating or preventing neurologic disorders and age-dependent cognitive decline. Neuroregenerative medicine can take great advantage by the deepening of our knowledge on the molecular mechanisms underlying the properties of neurotrophic factors. It is indeed an intriguing topic that a significant interplay between neurotrophic factors and various metals can modulate the outcome of neuronal recovery. This review is particularly focused on the roles of GDNF, BDNF and NGF in motoneuron survival and recovery from injuries and evaluates the therapeutic potential of various neurotrophic factors in neuronal regeneration. The key role of metal homeostasis/dyshomeostasis and metal interaction with neurotrophic factors on neuronal pathophysiology is also highlighted as a novel mechanism and potential target for neuronal recovery. The progress in mechanistic studies in the field of neurotrophic factor-mediated neuroprotection and neural regeneration, aiming at a complete understanding of integrated pathways, offers possibilities for the development of novel neuroregenerative therapeutic approaches.
Collapse
Affiliation(s)
- Vincenzo Giuseppe Nicoletti
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), Section of Medical Biochemistry, University of Catania, 95124 Catania, Italy; (V.G.N.); (D.C.)
| | - Krisztián Pajer
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary;
| | - Damiano Calcagno
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), Section of Medical Biochemistry, University of Catania, 95124 Catania, Italy; (V.G.N.); (D.C.)
| | - Gholam Pajenda
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Research Centre for Traumatology of the Austrian Workers, 1200 Vienna, Austria;
- Department for Trauma Surgery, Medical University Vienna, 1090 Vienna, Austria
| | - Antal Nógrádi
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary;
- Correspondence: ; Tel.: +36-6-234-2855
| |
Collapse
|
23
|
Wu MS, Li XJ, Liu CY, Xu Q, Huang JQ, Gu S, Chen JX. Effects of Histone Modification in Major Depressive Disorder. Curr Neuropharmacol 2022; 20:1261-1277. [PMID: 34551699 PMCID: PMC9881074 DOI: 10.2174/1570159x19666210922150043] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/26/2021] [Accepted: 09/21/2021] [Indexed: 11/22/2022] Open
Abstract
Major depressive disorder (MDD) is a disease associated with many factors; specifically, environmental, genetic, psychological, and biological factors play critical roles. Recent studies have demonstrated that histone modification may occur in the human brain in response to severely stressful events, resulting in transcriptional changes and the development of MDD. In this review, we discuss five different histone modifications, histone methylation, histone acetylation, histone phosphorylation, histone crotonylation and histone β-hydroxybutyrylation, and their relationships with MDD. The utility of histone deacetylase (HDAC) inhibitors (HDACis) for MDD treatment is also discussed. As a large number of MDD patients in China have been treated with traditional Chineses medicine (TCM), we also discuss some TCM therapies, such as Xiaoyaosan (XYS), and their effects on histone modification. In summary, targeting histone modification may be a new strategy for elucidating the mechanism of MDD and a new direction for MDD treatment.
Collapse
Affiliation(s)
- Man-Si Wu
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China;
| | - Xiao-Juan Li
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China;
| | - Chen-Yue Liu
- Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China;
| | - Qiuyue Xu
- Department of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China;
| | - Jun-Qing Huang
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China;
| | - Simeng Gu
- Department of Psychology, Jiangsu University Medical School, Zhenjiang, China
| | - Jia-Xu Chen
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China; ,Address correspondence to this author at the Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China; E-mail:
| |
Collapse
|
24
|
Truong TT, Bortolasci CC, Kidnapillai S, Spolding B, Panizzutti B, Liu ZS, Watmuff B, Kim JH, Dean OM, Richardson M, Berk M, Walder K. Common effects of bipolar disorder medications on expression quantitative trait loci genes. J Psychiatr Res 2022; 150:105-112. [PMID: 35366598 DOI: 10.1016/j.jpsychires.2022.03.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/23/2022] [Accepted: 03/21/2022] [Indexed: 10/18/2022]
Abstract
The molecular mechanism(s) underpinning the clinical efficacy of the current drugs for bipolar disorder (BD) are largely unknown. This study evaluated the transcriptional perturbations potentially playing roles in the therapeutic efficacy of four commonly prescribed psychotropic drugs used to treat BD. NT2-N cells were treated with lamotrigine, lithium, quetiapine, valproate or vehicle control for 24 h. Genome-wide mRNA expression was quantified by RNA-sequencing. Incorporating drug-induced gene expression profiles with BD-associated transcriptional changes from post-mortem brains, we identified potential therapeutic-relevant genes associated with both drug treatments and BD pathophysiology and focused on expression quantitative trait loci (eQTL) genes with genome-wide association with BD. Each eQTL gene was ranked based on its potential role in the therapeutic effect across multiple drugs. The expression of highest-ranked eQTL genes were measured by RT-qPCR to confirm their transcriptional changes observed in RNA-seq. We found 775 genes for which at least 2 drugs reversed expression levels relative to the differential expression in post-mortem brains. Pathway analysis identified enriched biological processes highlighting mitochondrial and endoplasmic reticulum function. Differential expression of SRPK2 and CHDH was confirmed by RT-qPCR following multiple-dose treatments. We pinpointed potential genes involved in the beneficial effects of drugs used for BD and their main associated biological pathways. CHDH, which encodes a mitochondrial protein, had a significant dose-responsive downregulation following treatment with increasing doses of quetiapine and lamotrigine, which in combination with the enriched mitochondrial pathways suggests potential therapeutic roles and demand more studies on mitochondrial involvement in BD to identify novel treatment targets.
Collapse
Affiliation(s)
- Trang Tt Truong
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, Australia.
| | - Chiara C Bortolasci
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, Australia
| | - Srisaiyini Kidnapillai
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, Australia
| | - Briana Spolding
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, Australia
| | - Bruna Panizzutti
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, Australia
| | - Zoe Sj Liu
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, Australia
| | - Brad Watmuff
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, Australia
| | - Jee Hyun Kim
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, Australia; Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Olivia M Dean
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, Australia; Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Mark Richardson
- Bioinformatics Core Research Facility (BCRF), Deakin University, Geelong, Australia
| | - Michael Berk
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, Australia; Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Ken Walder
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, Australia
| |
Collapse
|
25
|
Zhao XP, Li H, Dai RP. Neuroimmune crosstalk through brain-derived neurotrophic factor and its precursor pro-BDNF: New insights into mood disorders. World J Psychiatry 2022; 12:379-392. [PMID: 35433323 PMCID: PMC8968497 DOI: 10.5498/wjp.v12.i3.379] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 08/22/2021] [Accepted: 01/23/2022] [Indexed: 02/06/2023] Open
Abstract
Mood disorders are the most common mental disorders, affecting approximately 350 million people globally. Recent studies have shown that neuroimmune interaction regulates mood disorders. Brain-derived neurotrophic factor (BDNF) and its precursor pro-BDNF, are involved in the neuroimmune crosstalk during the development of mood disorders. BDNF is implicated in the pathophysiology of psychiatric and neurological disorders especially in antidepressant pharmacotherapy. In this review, we describe the functions of BDNF/pro-BDNF signaling in the central nervous system in the context of mood disorders. In addition, we summarize the developments for BDNF and pro-BDNF functions in mood disorders. This review aims to provide new insights into the impact of neuroimmune interaction on mood disorders and reveal a new basis for further development of diagnostic targets and mood disorders.
Collapse
Affiliation(s)
- Xiao-Pei Zhao
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Hui Li
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Ru-Ping Dai
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| |
Collapse
|
26
|
Gao L, Zhang Y, Sterling K, Song W. Brain-derived neurotrophic factor in Alzheimer's disease and its pharmaceutical potential. Transl Neurodegener 2022; 11:4. [PMID: 35090576 PMCID: PMC8796548 DOI: 10.1186/s40035-022-00279-0] [Citation(s) in RCA: 164] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 01/01/2022] [Indexed: 12/14/2022] Open
Abstract
Synaptic abnormalities are a cardinal feature of Alzheimer's disease (AD) that are known to arise as the disease progresses. A growing body of evidence suggests that pathological alterations to neuronal circuits and synapses may provide a mechanistic link between amyloid β (Aβ) and tau pathology and thus may serve as an obligatory relay of the cognitive impairment in AD. Brain-derived neurotrophic factors (BDNFs) play an important role in maintaining synaptic plasticity in learning and memory. Considering AD as a synaptic disorder, BDNF has attracted increasing attention as a potential diagnostic biomarker and a therapeutical molecule for AD. Although depletion of BDNF has been linked with Aβ accumulation, tau phosphorylation, neuroinflammation and neuronal apoptosis, the exact mechanisms underlying the effect of impaired BDNF signaling on AD are still unknown. Here, we present an overview of how BDNF genomic structure is connected to factors that regulate BDNF signaling. We then discuss the role of BDNF in AD and the potential of BDNF-targeting therapeutics for AD.
Collapse
Affiliation(s)
- Lina Gao
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, College of Pharmacy, Jining Medical University, Jining, 272067, Shandong, China
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Yun Zhang
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Keenan Sterling
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Weihong Song
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, College of Pharmacy, Jining Medical University, Jining, 272067, Shandong, China.
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and The Affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, 325001, Zhejiang, China.
| |
Collapse
|
27
|
Wang CS, Kavalali ET, Monteggia LM. BDNF signaling in context: From synaptic regulation to psychiatric disorders. Cell 2022; 185:62-76. [PMID: 34963057 PMCID: PMC8741740 DOI: 10.1016/j.cell.2021.12.003] [Citation(s) in RCA: 211] [Impact Index Per Article: 105.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/10/2021] [Accepted: 12/02/2021] [Indexed: 01/09/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) is a neuropeptide that plays numerous important roles in synaptic development and plasticity. While its importance in fundamental physiology is well established, studies of BDNF often produce conflicting and unclear results, and the scope of existing research makes the prospect of setting future directions daunting. In this review, we examine the importance of spatial and temporal factors on BDNF activity, particularly in processes such as synaptogenesis, Hebbian plasticity, homeostatic plasticity, and the treatment of psychiatric disorders. Understanding the fundamental physiology of when, where, and how BDNF acts and new approaches to control BDNF signaling in time and space can contribute to improved therapeutics and patient outcomes.
Collapse
Affiliation(s)
- Camille S Wang
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232-2050, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, USA
| | - Ege T Kavalali
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232-2050, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, USA
| | - Lisa M Monteggia
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232-2050, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, USA.
| |
Collapse
|
28
|
Chen PS, Tang LY, Chang HH. Roles of C-reactive protein polymorphisms and life event changes on cognitive function in bipolar patients receiving valproate. Int J Immunopathol Pharmacol 2022; 36:3946320221084835. [PMID: 35377256 PMCID: PMC8984865 DOI: 10.1177/03946320221084835] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Introduction Patients with bipolar disorder (BD) exhibit an inflamed condition that is
associated with metabolic disturbance and cognitive impairment. Whether
inflammation, represented by C-reactive protein (CRP), is causally
associated with BD and influences treatment outcome has not been
established. Methods We examined whether CRP is a causal factor for the risk of BD in drug-naïve,
depressed BD patients and investigated whether polymorphisms in
CRP and life event changes influence cognitive function
in BD patients receiving valproate (VPA) treatment. Results Our results showed that BD patients had significantly higher CRP levels and
worse cognitive function than the controls, while the frequencies of
CRP single nucleotide polymorphisms in BD patients and
in controls were not different. In addition, the life event scale score was
higher for BD patients than for controls. Furthermore, the genotypes of
CRP polymorphisms and the interactions between
polymorphisms of CRP and life event scale score had a
significant influence on cognitive performance in BD patients after 12 weeks
of VPA treatment. Conclusion Our study demonstrated the clinical utility of the application of functional
genetics in clarifying the interactions among CRP, life event stress, and BD
and suggested the important roles of CRP gene–environment
interactions in developing treatment strategies for BD.
Collapse
Affiliation(s)
- Po See Chen
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, 38026National Cheng Kung University, Tainan, Taiwan.,Institute of Behavioral Medicine, College of Medicine, 38026National Cheng Kung University, Tainan, Taiwan
| | - Li-Yi Tang
- Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, 38026National Cheng Kung University, Tainan, Taiwan
| | - Hui Hua Chang
- Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, 38026National Cheng Kung University, Tainan, Taiwan.,School of Pharmacy, College of Medicine, 38026National Cheng Kung University, Tainan, Taiwan.,Department of Pharmacy, National Cheng Kung University Hospital, College of Medicine, 38026National Cheng Kung University, Tainan, Taiwan.,Department of Pharmacy, National Cheng Kung University Hospital, Dou-Liou Branch, Yunlin, Taiwan
| |
Collapse
|
29
|
Pacholko AG, Bekar LK. Lithium orotate: A superior option for lithium therapy? Brain Behav 2021; 11:e2262. [PMID: 34196467 PMCID: PMC8413749 DOI: 10.1002/brb3.2262] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 06/08/2021] [Indexed: 01/23/2023] Open
Abstract
Bipolar disorder (BD) poses a significant public health concern, with roughly one-quarter of sufferers attempting suicide. BD is characterized by manic and depressive mood cycles, the recurrence of which can be effectively curtailed through lithium therapy. Unfortunately, the most frequently employed lithium salt, lithium carbonate (Li2 CO3 ), is associated with a host of adverse health outcomes following chronic use: these unwanted effects range from relatively minor inconveniences (e.g., polydipsia and polyuria) to potentially major complications (e.g., hypothyroidism and/or renal impairment). As these undesirable effects can limit patient compliance, an alternative lithium compound with a lesser toxicity profile would dramatically improve treatment efficacy and outcomes. Lithium orotate (LiC5 H3 N2 O4 ; henceforth referred to as LiOr), a compound largely abandoned since the late 1970s, may represent such an alternative. LiOr is proposed to cross the blood-brain barrier and enter cells more readily than Li2 CO3 , which will theoretically allow for reduced dosage requirements and ameliorated toxicity concerns. This review addresses the controversial history of LiOr, complete with discussions of experimental and clinical efficacy, putative mechanisms of action, adverse effects, and its potential future in therapy.
Collapse
Affiliation(s)
- Anthony G Pacholko
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Lane K Bekar
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
30
|
Modulation of Brain-Derived Neurotrophic Factor (BDNF) Signaling Pathway by Culinary Sage ( Salvia officinalis L.). Int J Mol Sci 2021; 22:ijms22147382. [PMID: 34299002 PMCID: PMC8303624 DOI: 10.3390/ijms22147382] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/22/2021] [Accepted: 06/26/2021] [Indexed: 01/19/2023] Open
Abstract
Culinary sage (Salvia officinalis L.) is a common spice plant in the mint family (Lamiaceae) well known for its distinctive culinary and traditional medicinal uses. Sage tea has been used traditionally as a brain-enhancing tonic and extracts from sage have been reported to have both cognitive and memory enhancing effects. Brain-derived neurotrophic factor (BDNF) is an endogenous signaling molecule involved in cognition and memory function. In this study, activity-guided fractionation employing preparative reverse-phase high performance liquid chromatography (RP-HPLC) of culinary sage extracts led to the discovery of benzyl 6-O-β-D-apiofuranosyl-β-D-glucoside (B6AG) as a natural product that upregulates transcription of neurotrophic factors in C6 glioma cells. Purified B6AG showed a moderate dose response, with upregulation of BDNF and with EC50 at 6.46 μM. To better understand the natural variation in culinary sage, B6AG was quantitated in the leaves of several commercial varieties by liquid chromatography-mass spectrometry (LC-MS). The level of B6AG in dried culinary sage was found to range from 334 ± 14 to 698 ± 65 μg/g. This study provided a foundation for future investigations, including quantitative inquiries on the distribution of B6AG within the different plant organs, explorations in optimizing post-harvest practices, and aid in the development of sage varieties with elevated levels of B6AG.
Collapse
|
31
|
Ribeiro ACR, Jahr FM, Hawkins E, Kronfol MM, Younis RM, McClay JL, Deshpande LS. Epigenetic histone acetylation and Bdnf dysregulation in the hippocampus of rats exposed to repeated, low-dose diisopropylfluorophosphate. Life Sci 2021; 281:119765. [PMID: 34186043 DOI: 10.1016/j.lfs.2021.119765] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/08/2021] [Accepted: 06/21/2021] [Indexed: 10/21/2022]
Abstract
AIMS Deployment-related exposures to organophosphate (OP) compounds are implicated for Gulf War Illness (GWI) development in First GW veterans. However, reasons for the persistence of GWI are not fully understood. Epigenetic modifications to chromatin are regulatory mechanisms that can adaptively or maladaptively respond to external stimuli. These include DNA methylation and histone acetylation. DNA methylation changes have been reported in GWI but the role of histone acetylation in GWI has been less explored, despite its importance as an epigenetic mechanism for neurological disorders. MAIN METHODS Male Sprague-Dawley rats were exposed to OP diisopropyl fluorophosphate (DFP, 0.5 mg/kg s.c., 5-d) and 6-m later brains were dissected for hippocampus. Western blotting, activity assays and chromatin immunoprecipitation (ChIP) were utilized for epigenetic analyses. Behavior was assessed using the Forced Swim Test (FST) and the Elevated Plus Maze (EPM). KEY FINDINGS We observed a significant upregulation in HDAC1 protein along with a significant increase in HDAC enzyme activity in the hippocampus of DFP rats. A locus-specific ChIP study revealed decreases in H3K9ac at the brain derived neurotrophic factor (Bdnf) promoter IV coupled with a significant decrease in BDNF protein in DFP rat hippocampus. Treatment with HDAC inhibitor valproic acid reduced HDAC activity and decreased the FST immobility time in DFP rats. SIGNIFICANCE Our research suggests that epigenetic alterations to histone acetylation pathways and decreased BDNF expression could represent novel mechanisms for GWI symptomatology and may provide new targets for developing effective drugs for GWI treatment.
Collapse
Affiliation(s)
- Ana C R Ribeiro
- Department of Neurology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Fay M Jahr
- Department of Pharmacotherapy & Outcome Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| | - Elisa Hawkins
- Department of Neurology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Mohamad M Kronfol
- Department of Pharmacotherapy & Outcome Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| | - Rabha M Younis
- Department of Pharmacotherapy & Outcome Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| | - Joseph L McClay
- Department of Pharmacotherapy & Outcome Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| | - Laxmikant S Deshpande
- Department of Neurology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA; Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
32
|
Razzaghi N, Fernandez-Gonzalez P, Mas-Sanchez A, Vila-Julià G, Perez JJ, Garriga P. Effect of Sodium Valproate on the Conformational Stability of the Visual G Protein-Coupled Receptor Rhodopsin. Molecules 2021; 26:molecules26103032. [PMID: 34069614 PMCID: PMC8160834 DOI: 10.3390/molecules26103032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/13/2021] [Accepted: 05/17/2021] [Indexed: 12/29/2022] Open
Abstract
Rhodopsin is the G protein-coupled receptor of rod photoreceptor cells that mediates vertebrate vision at low light intensities. Mutations in rhodopsin cause inherited retinal degenerative diseases such as retinitis pigmentosa. Several therapeutic strategies have attempted to address and counteract the deleterious effect of rhodopsin mutations on the conformation and function of this photoreceptor protein, but none has been successful in efficiently preventing retinal degeneration in humans. These approaches include, among others, the use of small molecules, known as pharmacological chaperones, that bind to the receptor stabilizing its proper folded conformation. Valproic acid, in its sodium valproate form, has been used as an anticonvulsant in epileptic patients and in the treatment of several psychiatric disorders. More recently, this compound has been tested as a potential therapeutic agent for the treatment of retinal degeneration associated with retinitis pigmentosa caused by rhodopsin mutations. We now report on the effect of sodium valproate on the conformational stability of heterologously expressed wild-type rhodopsin and a rhodopsin mutant, I307N, which has been shown to be an appropriate model for studying retinal degeneration in mice. We found no sign of enhanced stability for the dark inactive conformation of the I307N mutant. Furthermore, the photoactivated conformation of the mutant appears to be destabilized by sodium valproate as indicated by a faster decay of its active conformation. Therefore, our results support a destabilizing effect of sodium valproate on rhodopsin I307N mutant associated with retinal degeneration. These findings, at the molecular level, agree with recent clinical studies reporting negative effects of sodium valproate on the visual function of retinitis pigmentosa patients.
Collapse
Affiliation(s)
- Neda Razzaghi
- Grup de Biotecnologia Molecular i Industrial, Centre de Biotecnologia Molecular, Departament d’Enginyeria Química, Universitat Politècnica de Catalunya-Barcelona Tech, Edifici Gaia, Rambla de Sant Nebridi 22, 08222 Terrassa, Spain; (N.R.); (P.F.-G.); (A.M.-S.)
| | - Pol Fernandez-Gonzalez
- Grup de Biotecnologia Molecular i Industrial, Centre de Biotecnologia Molecular, Departament d’Enginyeria Química, Universitat Politècnica de Catalunya-Barcelona Tech, Edifici Gaia, Rambla de Sant Nebridi 22, 08222 Terrassa, Spain; (N.R.); (P.F.-G.); (A.M.-S.)
| | - Aina Mas-Sanchez
- Grup de Biotecnologia Molecular i Industrial, Centre de Biotecnologia Molecular, Departament d’Enginyeria Química, Universitat Politècnica de Catalunya-Barcelona Tech, Edifici Gaia, Rambla de Sant Nebridi 22, 08222 Terrassa, Spain; (N.R.); (P.F.-G.); (A.M.-S.)
| | - Guillem Vila-Julià
- Grup de Biotecnologia Molecular i Industrial, Centre de Biotecnologia Molecular, Departament d’Enginyeria Química, Universitat Politècnica de Catalunya-Barcelona Tech., Avinguda Diagonal, 647, 08028 Barcelona, Spain; (G.V.-J.); (J.J.P.)
| | - Juan Jesus Perez
- Grup de Biotecnologia Molecular i Industrial, Centre de Biotecnologia Molecular, Departament d’Enginyeria Química, Universitat Politècnica de Catalunya-Barcelona Tech., Avinguda Diagonal, 647, 08028 Barcelona, Spain; (G.V.-J.); (J.J.P.)
| | - Pere Garriga
- Grup de Biotecnologia Molecular i Industrial, Centre de Biotecnologia Molecular, Departament d’Enginyeria Química, Universitat Politècnica de Catalunya-Barcelona Tech, Edifici Gaia, Rambla de Sant Nebridi 22, 08222 Terrassa, Spain; (N.R.); (P.F.-G.); (A.M.-S.)
- Correspondence:
| |
Collapse
|
33
|
Jiang H, Xiao L, Jin K, Shao B. Estrogen administration attenuates post-stroke depression by enhancing CREB/BDNF/TrkB signaling in the rat hippocampus. Exp Ther Med 2021; 21:433. [PMID: 33747172 PMCID: PMC7967838 DOI: 10.3892/etm.2021.9850] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 10/09/2020] [Indexed: 12/27/2022] Open
Abstract
A previous study demonstrated that 17β-estradiol (E2), which is an antidepressant, can ameliorate post-stroke depression (PSD); however, the underlying mechanisms governing this remain largely unknown. Therefore, the present study developed a PSD model in rats, which was induced by left middle cerebral artery occlusion followed by exposure to chronic mild stress for 2 weeks. The results revealed that the activity of the cAMP response element-binding protein (CREB), a cellular transcription factor, and the associated brain-derived neurotrophic factor (BDNF)/tyrosine kinase B (TrkB) signaling were all attenuated in the hippocampus in PSD rats. The depression-like behaviors were significantly improved after treatment with E2, along with increased CREB and the BDNF/TrkB signaling activity. These results provide novel insight into the molecular basis of PSD, and suggest the potential involvement of CREB/BDNF/TrkB signaling in E2-mediated improvement of PSD in rats.
Collapse
Affiliation(s)
- Huigang Jiang
- Department of Neurology, Yiwu City Center Hospital, Wenzhou Medical University, Yiwu, Zhejiang 322000, P.R. China
| | - Li Xiao
- Department of Neurology, Shaoyang City Center Hospital, Shaoyang, Hunan 422000, P.R. China
| | - Kunlin Jin
- Department of Neurology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA
| | - Bei Shao
- Department of Neurology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
34
|
Petersen NA, Nielsen MØ, Coello K, Stanislaus S, Melbye S, Kjærstad HL, Sletved KSO, McIntyre RS, Frikke-Smith R, Vinberg M, Kessing LV. Brain-derived neurotrophic factor levels in newly diagnosed patients with bipolar disorder, their unaffected first-degree relatives and healthy controls. BJPsych Open 2021; 7:e55. [PMID: 33588978 PMCID: PMC8058924 DOI: 10.1192/bjo.2021.9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF), which facilitates neuroplasticity and synaptogenesis, may be decreased in bipolar disorder, but has not been systematically investigated in people with newly diagnosed bipolar disorder and unaffected first-degree relatives. AIMS To compare BDNF levels in patients with newly diagnosed bipolar disorder, their unaffected first-degree relatives and healthy controls. METHOD The study investigated plasma BDNF levels in patients (n = 371) with newly diagnosed bipolar disorder, their unaffected first-degree relatives (n = 98) and healthy controls (n = 200) using enzyme-linked immunosorbent assay. We further investigated associations between BDNF levels and illness-related variables and medication status. RESULTS BDNF levels were found to be 22.0% (95% CI 1.107-1.343) higher in patients with bipolar disorder compared with healthy controls (P < 0.001) and 15.6% higher in unaffected first-degree relatives compared with healthy controls (95% CI 1.007-1.327, P = 0.04), when adjusting for age and gender. Further, BDNF levels were positively associated with duration of illness at a trend level (P = 0.05), age (P = 0.001) and use of anti-epileptic medication (P = 0.05). CONCLUSIONS These findings suggest that BDNF levels are not decreased in the early stages of bipolar disorder and in unaffected first-degree relatives contrasting with prior findings during later stages of the illness.
Collapse
Affiliation(s)
- Nanna Aagaard Petersen
- Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Centre Copenhagen, Rigshospitalet, Denmark
| | - Marc Østergaard Nielsen
- Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Centre Copenhagen, Rigshospitalet, Denmark
| | - Klara Coello
- Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Centre Copenhagen, Rigshospitalet, Denmark
| | - Sharleny Stanislaus
- Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Centre Copenhagen, Rigshospitalet, Denmark
| | - Sigurd Melbye
- Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Centre Copenhagen, Rigshospitalet, Denmark
| | - Hanne Lie Kjærstad
- Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Centre Copenhagen, Rigshospitalet, Denmark
| | | | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
| | - Ruth Frikke-Smith
- Department of Clinical Biochemistry, Centre of Diagnostic Investigation, Rigshospitalet, Denmark
| | - Maj Vinberg
- Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Centre Copenhagen, Rigshospitalet, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; and Psychiatric Research Unit, Psychiatric Centre North Zealand, Hillerød, Denmark
| | - Lars Vedel Kessing
- Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Centre Copenhagen, Rigshospitalet, Denmark; and Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
35
|
Chen J, Wang X, Hu J, Du J, Dordoe C, Zhou Q, Huang W, Guo R, Han F, Guo K, Ye S, Lin L, Li X. FGF20 Protected Against BBB Disruption After Traumatic Brain Injury by Upregulating Junction Protein Expression and Inhibiting the Inflammatory Response. Front Pharmacol 2021; 11:590669. [PMID: 33568994 PMCID: PMC7868342 DOI: 10.3389/fphar.2020.590669] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/14/2020] [Indexed: 12/11/2022] Open
Abstract
Disruption of the blood-brain barrier (BBB) and the cerebral inflammatory response occurring after traumatic brain injury (TBI) facilitate further brain damage, which leads to long-term complications of TBI. Fibroblast growth factor 20 (FGF20), a neurotrophic factor, plays important roles in brain development and neuronal homeostasis. The aim of the current study was to assess the protective effects of FGF20 on TBI via BBB maintenance. In the present study, recombinant human FGF20 (rhFGF20) reduced neurofunctional deficits, brain edema, Evans blue extravasation and neuroinflammation in a TBI mouse model. In an in vitro TNF-α-induced human brain microvascular endothelial cell (HBMEC) model of BBB disruption, rhFGF20 reduced paracellular permeability and increased trans-endothelial electrical resistance (TEER). Both in the TBI mouse model and in vitro, rhFGF20 increased the expression of proteins composing in BBB-associated tight junctions (TJs) and adherens junctions (AJs), and decreased the inflammatory response, which protected the BBB integrity. Notably, rhFGF20 preserved BBB function by activating the AKT/GSK3β pathway and inhibited the inflammatory response by regulating the JNK/NFκB pathway. Thus, FGF20 is a potential candidate treatment for TBI that protects the BBB by upregulating junction protein expression and inhibiting the inflammatory response.
Collapse
Affiliation(s)
- Jun Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xue Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jian Hu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jingting Du
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Confidence Dordoe
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Qiulin Zhou
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wenting Huang
- School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ruili Guo
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Fanyi Han
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Kaiming Guo
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shasha Ye
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Li Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Research Units of Clinical Translation of Cell Growth Factors and Diseases Research, Chinese Academy of Medical Science, Wenzhou, China
| |
Collapse
|
36
|
Lack of Autophagy Induction by Lithium Decreases Neuroprotective Effects in the Striatum of Aged Rats. Pharmaceutics 2021; 13:pharmaceutics13020135. [PMID: 33494241 PMCID: PMC7909773 DOI: 10.3390/pharmaceutics13020135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/14/2021] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
The pharmacological modulation of autophagy is considered a promising neuroprotective strategy. While it has been postulated that lithium regulates this cellular process, the age-related effects have not been fully elucidated. Here, we evaluated lithium-mediated neuroprotective effects in young and aged striatum. After determining the optimal experimental conditions for inducing autophagy in loco with lithium carbonate (Li2CO3), we measured cell viability, reactive oxygen species (ROS) generation and oxygen consumption with rat brain striatal slices from young and aged animals. In the young striatum, Li2CO3 increased tissue viability and decreased ROS generation. These positive effects were accompanied by enhanced levels of LC3-II, LAMP 1, Ambra 1 and Beclin-1 expression. In the aged striatum, Li2CO3 reduced the autophagic flux and increased the basal oxygen consumption rate. Ultrastructural changes in the striatum of aged rats that consumed Li2CO3 for 30 days included electrondense mitochondria with disarranged cristae and reduced normal mitochondria and lysosomes area. Our data show that the striatum from younger animals benefits from lithium-mediated neuroprotection, while the striatum of older rats does not. These findings should be considered when developing neuroprotective strategies involving the induction of autophagy in aging.
Collapse
|
37
|
Albeely AM, Williams OOF, Perreault ML. GSK-3β Disrupts Neuronal Oscillatory Function to Inhibit Learning and Memory in Male Rats. Cell Mol Neurobiol 2021; 42:1341-1353. [PMID: 33392916 DOI: 10.1007/s10571-020-01020-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/27/2020] [Indexed: 12/25/2022]
Abstract
Alterations in glycogen synthase kinase-3β (GSK-3β) activity have been implicated in disorders of cognitive impairment, including Alzheimer's disease and schizophrenia. Cognitive dysfunction is also characterized by the dysregulation of neuronal oscillatory activity, macroscopic electrical rhythms in brain that are critical to systems communication. A direct functional relationship between GSK-3β and neuronal oscillations has not been elucidated. Therefore, in the present study, using an adeno-associated viral vector containing a persistently active mutant form of GSK-3β, GSK-3β(S9A), the impact of elevated kinase activity in prefrontal cortex (PFC) or ventral hippocampus (vHIP) of rats on neuronal oscillatory activity was evaluated. GSK-3β(S9A)-induced changes in learning and memory were also assessed and the phosphorylation status of tau protein, a substrate of GSK-3β, examined. It was demonstrated that increasing GSK-3β(S9A) activity in either the PFC or vHIP had similar effects on neuronal oscillatory activity, enhancing theta and/or gamma spectral power in one or both regions. Increasing PFC GSK-3β(S9A) activity additionally suppressed high gamma PFC-vHIP coherence. These changes were accompanied by deficits in recognition memory, spatial learning, and/or reversal learning. Elevated pathogenic tau phosphorylation was also evident in regions where GSK-3β(S9A) activity was upregulated. The neurophysiological and learning and memory deficits induced by GSK-3β(S9A) suggest that aberrant GSK-3β signalling may not only play an early role in cognitive decline in Alzheimer's disease but may also have a more central involvement in disorders of cognitive dysfunction through the regulation of neurophysiological network function.
Collapse
Affiliation(s)
- Abdalla M Albeely
- Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd. E, Guelph, ON, N1G 2W1, Canada.,Collaborative Neuroscience Program, University of Guelph, 50 Stone Rd. E, Guelph, ON, Canada
| | - Olivia O F Williams
- Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd. E, Guelph, ON, N1G 2W1, Canada
| | - Melissa L Perreault
- Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd. E, Guelph, ON, N1G 2W1, Canada. .,Collaborative Neuroscience Program, University of Guelph, 50 Stone Rd. E, Guelph, ON, Canada.
| |
Collapse
|
38
|
Is There Justification to Treat Neurodegenerative Disorders by Repurposing Drugs? The Case of Alzheimer's Disease, Lithium, and Autophagy. Int J Mol Sci 2020; 22:ijms22010189. [PMID: 33375448 PMCID: PMC7795249 DOI: 10.3390/ijms22010189] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/16/2020] [Accepted: 12/23/2020] [Indexed: 12/11/2022] Open
Abstract
Lithium is the prototype mood-stabilizer used for acute and long-term treatment of bipolar disorder. Cumulated translational research of lithium indicated the drug's neuroprotective characteristics and, thereby, has raised the option of repurposing it as a drug for neurodegenerative diseases. Lithium's neuroprotective properties rely on its modulation of homeostatic mechanisms such as inflammation, mitochondrial function, oxidative stress, autophagy, and apoptosis. This myriad of intracellular responses are, possibly, consequences of the drug's inhibition of the enzymes inositol-monophosphatase (IMPase) and glycogen-synthase-kinase (GSK)-3. Here we review lithium's neurobiological properties as evidenced by its neurotrophic and neuroprotective properties, as well as translational studies in cells in culture, in animal models of Alzheimer's disease (AD) and in patients, discussing the rationale for the drug's use in the treatment of AD.
Collapse
|
39
|
Masnata M, Salem S, de Rus Jacquet A, Anwer M, Cicchetti F. Targeting Tau to Treat Clinical Features of Huntington's Disease. Front Neurol 2020; 11:580732. [PMID: 33329322 PMCID: PMC7710872 DOI: 10.3389/fneur.2020.580732] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/17/2020] [Indexed: 12/16/2022] Open
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder characterized by severe motor, cognitive and psychiatric impairments. While motor deficits often confirm diagnosis, cognitive dysfunctions usually manifest early in the disease process and are consistently ranked among the leading factors that impact the patients' quality of life. The genetic component of HD, a mutation in the huntingtin (HTT) gene, is traditionally presented as the main contributor to disease pathology. However, accumulating evidence suggests the implication of the microtubule-associated tau protein to the pathogenesis and therefore, proposes an alternative conceptual framework where tau and mutant huntingtin (mHTT) act conjointly to drive neurodegeneration and cognitive dysfunction. This perspective on disease etiology offers new avenues to design therapeutic interventions and could leverage decades of research on Alzheimer's disease (AD) and other tauopathies to rapidly advance drug discovery. In this mini review, we examine the breadth of tau-targeting treatments currently tested in the preclinical and clinical settings for AD and other tauopathies, and discuss the potential application of these strategies to HD.
Collapse
Affiliation(s)
- Maria Masnata
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC, Canada.,Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, Canada
| | - Shireen Salem
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC, Canada.,Département de Médecine Moléculaire, Université Laval, Québec, QC, Canada
| | - Aurelie de Rus Jacquet
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC, Canada.,Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, Canada
| | - Mehwish Anwer
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC, Canada.,Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, Canada
| | - Francesca Cicchetti
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC, Canada.,Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, Canada.,Département de Médecine Moléculaire, Université Laval, Québec, QC, Canada
| |
Collapse
|
40
|
HDAC inhibition ameliorates cone survival in retinitis pigmentosa mice. Cell Death Differ 2020; 28:1317-1332. [PMID: 33159184 PMCID: PMC8026998 DOI: 10.1038/s41418-020-00653-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/12/2020] [Accepted: 10/15/2020] [Indexed: 12/27/2022] Open
Abstract
Cone photoreceptor cell death in inherited retinal diseases, such as Retinitis Pigmentosa (RP), leads to the loss of high acuity and color vision and, ultimately to blindness. In RP, a vast number of mutations perturb the structure and function of rod photoreceptors, while cones remain initially unaffected. Extensive rod loss in advanced stages of the disease triggers cone death by a mechanism that is still largely unknown. Here, we show that secondary cone cell death in animal models for RP is associated with increased activity of histone deacetylates (HDACs). A single intravitreal injection of an HDAC inhibitor at late stages of the disease, when the majority of rods have already degenerated, was sufficient to delay cone death and support long-term cone survival in two mouse models for RP, affected by mutations in the phosphodiesterase 6b gene. Moreover, the surviving cones remained light-sensitive, leading to an improvement in visual function. RNA-seq analysis of protected cones demonstrated that HDAC inhibition initiated multi-level protection via regulation of different pro-survival pathways, including MAPK, PI3K-Akt, and autophagy. This study suggests a unique opportunity for targeted pharmacological protection of secondary dying cones by HDAC inhibition and creates hope to maintain vision in RP patients even in advanced disease stages.
Collapse
|
41
|
Mesocortical BDNF signaling mediates antidepressive-like effects of lithium. Neuropsychopharmacology 2020; 45:1557-1566. [PMID: 32428928 PMCID: PMC7360776 DOI: 10.1038/s41386-020-0713-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/30/2020] [Accepted: 05/12/2020] [Indexed: 12/16/2022]
Abstract
Lithium has been used to treat major depressive disorder, yet the neural circuit mechanisms underlying this therapeutic effect remain unknown. Here, we demonstrated that the ventral tegmental area (VTA) dopamine (DA) neurons that project to the medial prefrontal cortex (mPFC), but not to nucleus accumbens (NAc), contributed to the antidepressive-like effects of lithium. Projection-specific electrophysiological recordings revealed that high concentrations of lithium increased firing rates in mPFC-, but not NAc-, projecting VTA DA neurons in mice treated with chronic unpredictable mild stress (CMS). In parallel, chronic administration of high-dose lithium in CMS mice restored the firing properties of mPFC-projecting DA neurons, and also rescued CMS-induced depressive-like behaviors. Nevertheless, chronic lithium treatment was insufficient to change the basal firing rates in NAc-projecting VTA DA neurons. Furthermore, chemogenetic activation of mPFC-, but not NAc-, projecting VTA DA neurons mimicked the antidepressive-like effects of lithium in CMS mice. Chemogenetic downregulation of VTA-mPFC DA neurons' firing activity abolished the antidepressive-like effects of lithium in CMS mice. Finally, we found that the antidepressant-like effects induced by high-dose lithium were mediated by BNDF signaling in the mesocortical DA circuit. Together, these results demonstrated the role of mesocortical DA projection in antidepressive-like effects of lithium and established a circuit foundation for lithium-based antidepressive treatment.
Collapse
|
42
|
Swann OG, Kilpatrick M, Breslin M, Oddy WH. Dietary fiber and its associations with depression and inflammation. Nutr Rev 2020; 78:394-411. [PMID: 31750916 DOI: 10.1093/nutrit/nuz072] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Dietary fiber is a crucial component of a healthy diet, with benefits that can be attributed to processes in the gut microbiota and the resulting by-products. Observational studies support associations between dietary fiber intake and depression and inflammation, but the potential mechanisms are poorly understood. This review examines evidence of the effects of dietary fiber on depression and inflammation and considers plausible mechanisms linking dietary fiber and depression, including microbiota-driven modification of gene expression and increased production of neurotransmitters. Additionally, inflammation may mediate the relationship between dietary fiber intake and depression. A high-fiber diet potentially lowers inflammation by modifying both the pH and the permeability of the gut. The resultant reduction in inflammatory compounds may alter neurotransmitter concentrations to reduce symptoms of depression. Further research into the link between dietary fiber intake and inflammation and depression is essential, as findings could potentially provide guidance for improvement in or prevention of inflammatory and depressive disorders.
Collapse
Affiliation(s)
- Olivia G Swann
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Michelle Kilpatrick
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Monique Breslin
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Wendy H Oddy
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
43
|
Impact of a Histone Deacetylase Inhibitor-Trichostatin A on Neurogenesis after Hypoxia-Ischemia in Immature Rats. Int J Mol Sci 2020; 21:ijms21113808. [PMID: 32471267 PMCID: PMC7312253 DOI: 10.3390/ijms21113808] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/14/2020] [Accepted: 05/26/2020] [Indexed: 12/22/2022] Open
Abstract
Hypoxia-ischemia (HI) in the neonatal brain frequently results in neurologic impairments, including cognitive disability. Unfortunately, there are currently no known treatment options to minimize ischemia-induced neural damage. We previously showed the neuroprotective/neurogenic potential of a histone deacetylase inhibitor (HDACi), sodium butyrate (SB), in a neonatal HI rat pup model. The aim of the present study was to examine the capacity of another HDACi—Trichostatin A (TSA)—to stimulate neurogenesis in the subgranular zone of the hippocampus. We also assessed some of the cellular/molecular processes that could be involved in the action of TSA, including the expression of neurotrophic factors (glial cell line-derived neurotrophic factor (GDNF), nerve growth factor (NGF), and brain-derived neurotrophic factor (BDNF)) as well as the TrkB receptor and its downstream signalling substrate— cAMP response element-binding protein (CREB). Seven-day-old rat pups were subjected to unilateral carotid artery ligation followed by hypoxia for 1 h. TSA was administered directly after the insult (0.2 mg/kg body weight). The study demonstrated that treatment with TSA restored the reduced by hypoxia-ischemia number of immature neurons (neuroblasts, BrdU/DCX-positive) as well as the number of oligodendrocyte progenitors (BrdU/NG2+) in the dentate gyrus of the ipsilateral damaged hemisphere. However, new generated cells did not develop the more mature phenotypes. Moreover, the administration of TSA stimulated the expression of BDNF and increased the activation of the TrkB receptor. These results suggest that BDNF-TrkB signalling pathways may contribute to the effects of TSA after neonatal hypoxic-ischemic injury.
Collapse
|
44
|
The early overgrowth theory of autism spectrum disorder: Insight into convergent mechanisms from valproic acid exposure and translational models. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020. [PMID: 32711813 DOI: 10.1016/bs.pmbts.2020.04.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
The development of new approaches for the clinical management of autism spectrum disorder (ASD) can only be realized through a better understanding of the neurobiological changes associated with ASD. One strategy for gaining deeper insight into the neurobiological mechanisms associated with ASD is to identify converging pathogenic processes associated with human idiopathic clinicopathology that are conserved in translational models of ASD. In this chapter, we first present the early overgrowth theory of ASD. Second, we introduce valproic acid (VPA), one of the most robust and well-known environmental risk factors associated with ASD, and we summarize the rapidly growing body of animal research literature using VPA as an ASD translational model. Lastly, we will detail the mechanisms of action of VPA and its impact on functional neural systems, as well as discuss future research directions that could have a lasting impact on the field.
Collapse
|
45
|
Park J, Cheon W, Kim K. Effects of Long-Term Endurance Exercise and Lithium Treatment on Neuroprotective Factors in Hippocampus of Obese Rats. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17093317. [PMID: 32397675 PMCID: PMC7246857 DOI: 10.3390/ijerph17093317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 01/03/2023]
Abstract
To investigate the effects of long-term lithium treatment and low intensity endurance exercise on brain-derived neurotrophic factor (BDNF) expression and glycogen synthase kinase 3 beta (GSK3β) activity in the hippocampus of obese rats. Fifty 10-week-old male Sprague-Dawley rats were selected. There was a control group of 10 rats (chow control group) while the other forty rats were fed on a high-fat diet for eight weeks to induce obesity. Rats were then assigned into four random groups. The rats were given 10 mg/kg lithium chloride (LiCl) dissolved in 1 mL sterile distilled water once a day, 5 times a week. The rats did 20 min of treadmill walking with an exercise intensity of 40% maximal oxygen uptake (VO2 max) (12 m/min, slope 0%). This was performed for 20 min a day, 3 days a week. Twelve weeks of lithium treatment or endurance exercise significantly reduced body weight and body fat mass in obese rats, without showing additive effects when the treatments were given in parallel or significant toxic responses in alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels in blood and kidney and liver tissues. BDNF expression in the hippocampus was significantly increased both in exercise and lithium groups with synergistic effects found in the group where both exercise and lithium treatments were given in parallel. On the other hand, the decrease in GSK3β activity was shown only in the lithium treatment group, without showing additive effects when the treatments were given in parallel. Lithium and low-intensity endurance exercise for 12 weeks increased the expression of BDNF, a neuroprotective factor in the hippocampus of obese mice. Lithium treatment alone inhibited the activity of GSK3β. This can be interpreted as a positive indication of applicability of the two factors in the prevention of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jusik Park
- Department of Taekwondo, College of Physical Education, Keimyung University, Daegu 42601, Korea;
| | - Wookwang Cheon
- Department of Physical Education, College of Physical Education, Keimyung University, Daegu 42601, Korea;
| | - Kijin Kim
- Department of Physical Education, College of Physical Education, Keimyung University, Daegu 42601, Korea;
- Correspondence: ; Tel.: +82-53-580-5256
| |
Collapse
|
46
|
Manduca JD, Thériault RK, Perreault ML. Glycogen synthase kinase-3: The missing link to aberrant circuit function in disorders of cognitive dysfunction? Pharmacol Res 2020; 157:104819. [PMID: 32305493 DOI: 10.1016/j.phrs.2020.104819] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/10/2020] [Accepted: 04/07/2020] [Indexed: 12/15/2022]
Abstract
Elevated GSK-3 activity has been implicated in cognitive dysfunction associated with various disorders including Alzheimer's disease, schizophrenia, type 2 diabetes, traumatic brain injury, major depressive disorder and bipolar disorder. Further, aberrant neural oscillatory activity in, and between, cortical regions and the hippocampus is consistently present within these same cognitive disorders. In this review, we will put forth the idea that increased GSK-3 activity serves as a pathological convergence point across cognitive disorders, inducing similar consequent impacts on downstream signaling mechanisms implicated in the maintenance of processes critical to brain systems communication and normal cognitive functioning. In this regard we suggest that increased activation of GSK-3 and neuronal oscillatory dysfunction are early pathological changes that may be functionally linked. Mechanistic commonalities between these disorders of cognitive dysfunction will be discussed and potential downstream targets of GSK-3 that may contribute to neuronal oscillatory dysfunction identified.
Collapse
Affiliation(s)
- Joshua D Manduca
- Department of Molecular and Cellular Biology, University of Guelph, ON, Canada
| | | | - Melissa L Perreault
- Department of Molecular and Cellular Biology, University of Guelph, ON, Canada.
| |
Collapse
|
47
|
Sial OK, Parise EM, Parise LF, Gnecco T, Bolaños-Guzmán CA. Ketamine: The final frontier or another depressing end? Behav Brain Res 2020; 383:112508. [PMID: 32017978 PMCID: PMC7127859 DOI: 10.1016/j.bbr.2020.112508] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/15/2020] [Accepted: 01/23/2020] [Indexed: 12/12/2022]
Abstract
Two decades ago, the observation of a rapid and sustained antidepressant response after ketamine administration provided an exciting new avenue in the search for more effective therapeutics for the treatment of clinical depression. Research elucidating the mechanism(s) underlying ketamine's antidepressant properties has led to the development of several hypotheses, including that of disinhibition of excitatory glutamate neurons via blockade of N-methyl-d-aspartate (NMDA) receptors. Although the prominent understanding has been that ketamine's mode of action is mediated solely via the NMDA receptor, this view has been challenged by reports implicating other glutamate receptors such as AMPA, and other neurotransmitter systems such as serotonin and opioids in the antidepressant response. The recent approval of esketamine (Spravato™) for the treatment of depression has sparked a resurgence of interest for a deeper understanding of the mechanism(s) underlying ketamine's actions and safe therapeutic use. This review aims to present our current knowledge on both NMDA and non-NMDA mechanisms implicated in ketamine's response, and addresses the controversy surrounding the antidepressant role and potency of its stereoisomers and metabolites. There is much that remains to be known about our understanding of ketamine's antidepressant properties; and although the arrival of esketamine has been received with great enthusiasm, it is now more important than ever that its mechanisms of action be fully delineated, and both the short- and long-term neurobiological/functional consequences of its treatment be thoroughly characterized.
Collapse
MESH Headings
- Antidepressive Agents/pharmacology
- Antidepressive Agents/therapeutic use
- Depressive Disorder, Major/drug therapy
- Depressive Disorder, Treatment-Resistant/drug therapy
- Dopamine Plasma Membrane Transport Proteins/drug effects
- Excitatory Amino Acid Antagonists/pharmacology
- Excitatory Amino Acid Antagonists/therapeutic use
- Humans
- Ketamine/pharmacology
- Ketamine/therapeutic use
- Norepinephrine Plasma Membrane Transport Proteins/drug effects
- Receptor, Muscarinic M1/drug effects
- Receptors, AMPA/drug effects
- Receptors, Dopamine D2/drug effects
- Receptors, N-Methyl-D-Aspartate/drug effects
- Receptors, Opioid, delta/drug effects
- Receptors, Opioid, kappa/drug effects
- Receptors, Opioid, mu/drug effects
- Receptors, Serotonin, 5-HT3/drug effects
- Receptors, sigma/drug effects
- Serotonin Plasma Membrane Transport Proteins/drug effects
Collapse
Affiliation(s)
- Omar K Sial
- Texas A&M University: Department of Psychological and Brain Sciences, 4325 TAMU, College Station, TX, 77843, USA
| | - Eric M Parise
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY, 10029, USA
| | - Lyonna F Parise
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY, 10029, USA
| | - Tamara Gnecco
- Texas A&M University: Department of Psychological and Brain Sciences, 4325 TAMU, College Station, TX, 77843, USA
| | - Carlos A Bolaños-Guzmán
- Texas A&M University: Department of Psychological and Brain Sciences, 4325 TAMU, College Station, TX, 77843, USA.
| |
Collapse
|
48
|
Magno LAV, Tenza-Ferrer H, Collodetti M, Nicolau EDS, Khlghatyan J, Del'Guidice T, Romano-Silva MA, Beaulieu JM. Contribution of neuronal calcium sensor 1 (Ncs-1) to anxiolytic-like and social behavior mediated by valproate and Gsk3 inhibition. Sci Rep 2020; 10:4566. [PMID: 32165725 PMCID: PMC7067888 DOI: 10.1038/s41598-020-61248-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 02/24/2020] [Indexed: 12/20/2022] Open
Abstract
Peripheral biomarker and post-mortem brains studies have shown alterations of neuronal calcium sensor 1 (Ncs-1) expression in people with bipolar disorder or schizophrenia. However, its engagement by psychiatric medications and potential contribution to behavioral regulation remains elusive. We investigated the effect on Ncs-1 expression of valproic acid (VPA), a mood stabilizer used for the management of bipolar disorder. Treatment with VPA induced Ncs-1 gene expression in cell line while chronic administration of this drug to mice increased both Ncs-1 protein and mRNA levels in the mouse frontal cortex. Inhibition of histone deacetylases (HDACs), a known biochemical effect of VPA, did not alter the expression of Ncs-1. In contrast, pharmacological inhibition or genetic downregulation of glycogen synthase kinase 3β (Gsk3β) increased Ncs-1 expression, whereas overexpression of a constitutively active Gsk3β had the opposite effect. Moreover, adeno-associated virus-mediated Ncs-1 overexpression in mouse frontal cortex caused responses similar to those elicited by VPA or lithium in tests evaluating social and mood-related behaviors. These findings indicate that VPA increases frontal cortex Ncs-1 gene expression as a result of Gsk3 inhibition. Furthermore, behavioral changes induced by Ncs-1 overexpression support a contribution of this mechanism in the regulation of behavior by VPA and potentially other psychoactive medications inhibiting Gsk3 activity.
Collapse
Affiliation(s)
- Luiz Alexandre Viana Magno
- Centro de Tecnologia em Medicina Molecular, Belo Horizonte, Brazil.,Departamento de Saúde Mental, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, CEP, 30130-100, Brazil.,Department of Psychiatry and Neuroscience, Laval University, Québec, Canada
| | - Helia Tenza-Ferrer
- Centro de Tecnologia em Medicina Molecular, Belo Horizonte, Brazil.,Departamento de Saúde Mental, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, CEP, 30130-100, Brazil
| | - Mélcar Collodetti
- Centro de Tecnologia em Medicina Molecular, Belo Horizonte, Brazil.,Departamento de Saúde Mental, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, CEP, 30130-100, Brazil
| | - Eduardo de Souza Nicolau
- Centro de Tecnologia em Medicina Molecular, Belo Horizonte, Brazil.,Departamento de Saúde Mental, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, CEP, 30130-100, Brazil
| | - Jivan Khlghatyan
- Department of Psychiatry and Neuroscience, Laval University, Québec, Canada.,Department of Pharmacology & Toxicology, University of Toronto, Toronto, Canada
| | - Thomas Del'Guidice
- Department of Psychiatry and Neuroscience, Laval University, Québec, Canada.,Feldan Therapeutics, Québec City, Canada
| | - Marco Aurélio Romano-Silva
- Centro de Tecnologia em Medicina Molecular, Belo Horizonte, Brazil. .,Departamento de Saúde Mental, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, CEP, 30130-100, Brazil.
| | - Jean Martin Beaulieu
- Department of Psychiatry and Neuroscience, Laval University, Québec, Canada. .,Department of Pharmacology & Toxicology, University of Toronto, Toronto, Canada.
| |
Collapse
|
49
|
Li M, Xia M, Chen W, Wang J, Yin Y, Guo C, Li C, Tang X, Zhao H, Tan Q, Chen Y, Jia Z, Liu X, Feng H. Lithium treatment mitigates white matter injury after intracerebral hemorrhage through brain-derived neurotrophic factor signaling in mice. Transl Res 2020; 217:61-74. [PMID: 31951826 DOI: 10.1016/j.trsl.2019.12.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 12/20/2019] [Accepted: 12/20/2019] [Indexed: 01/04/2023]
Abstract
Intracerebral hemorrhage (ICH), a subtype of stroke with high morbidity and mortality, occurs mainly in the basal ganglia and causes white matter injury (WMI), resulting in severe motor dysfunction and poor prognosis in patients. The preservation of the white matter around the hematoma is crucial for motor function recovery, but there is currently no effective treatment for WMI following ICH. Lithium has been widely used for the treatment of bipolar disorder for decades. Although the protective effects of lithium on neurodegenerative diseases and cerebral trauma have been studied in recent years, whether it can be used to alleviate WMI after ICH remains to be researched. The results of this study revealed that ICH caused significant functional and pathological abnormalities in mice. After LiCl was administered to mice with ICH, behavioural performance and electrophysiological functions were improved and ICH-induced white matter pathological injury, including myelin sheath and axonal degeneration, was ameliorated. Furthermore, LiCl treatment decreased the death of mature oligodendrocytes (OLGs) in ICH mice, which may have been attributed to the enhanced expression of brain-derived neurotrophic factor (BDNF) regulated by the LiCl-induced inhibition of glycogen synthase kinase-3β (GSK-3β). The decreased death of OLGs was closely associated with decreased destruction of the myelin sheath and alleviated degradation of the axons. In summary, this study suggests that the protective effect of lithium on WMI after ICH might be related to an increased level of BDNF and that LiCl treatment may be a potential therapeutic method to palliate WMI after ICH.
Collapse
Affiliation(s)
- Mingxi Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Min Xia
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Weixiang Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Jie Wang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Yi Yin
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Chao Guo
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Chengcheng Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Xiaoqin Tang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Hengli Zhao
- Department of Neurology, The Second Medical Central, Chinese PLA (People's Liberation Army) General Hospital, Beijing, PR China
| | - Qiang Tan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Yujie Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China; State Key Laboratory of Trauma, Burn, and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, PR China; Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Zhengcai Jia
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Xin Liu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China; Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China.
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China; State Key Laboratory of Trauma, Burn, and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, PR China; Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China.
| |
Collapse
|
50
|
Suga Y, Yoshimoto K, Numata S, Shimodera S, Takamura S, Kamimura N, Sawada K, Kazui H, Ohmori T, Morinobu S. Structural variation in the glycogen synthase kinase 3β and brain-derived neurotrophic factor genes in Japanese patients with bipolar disorders. Neuropsychopharmacol Rep 2019; 40:46-51. [PMID: 31769621 PMCID: PMC7292225 DOI: 10.1002/npr2.12083] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 10/20/2019] [Accepted: 10/21/2019] [Indexed: 01/06/2023] Open
Abstract
Background Lithium is the first‐line drug for the treatment of bipolar disorders (BDs); however, not all patients responded. Glycogen synthase kinase (GSK) 3β and brain‐derived neurotrophic factor (BDNF) play a role in the therapeutic action of lithium. Since structural variations were reported in these genes, it is possible that these genomic variations may be involved in the therapeutic responses to lithium. Method Fifty patients with BDs and 50 healthy subjects (mean age 55.0 ± 15.0 years; M/F 19/31) participated. We examined structural variation of the GSK3β and BDNF genes by real‐time PCR. We examined the influence of structural variation of these genes on the therapeutic responses to lithium and the occurrence of antidepressant‐emergent affective switch (AEAS). The efficacy of lithium was assessed using the Alda scale, and AEAS was evaluated using Young Mania Rating Scale. Results Although we examined structural variations within intron II and VII of the GSK3® gene and from the end of exon IV to intron IV and within exon IX of the BDNF gene, no structural variation was found in BDs. Whereas 5 of 50 patients exhibited three copies of the genomic region within exon IV of the BDNF gene, all healthy subjects had two copies. No difference in the therapeutic efficacy of lithium was found between patients with three and two copies. No difference in the occurrence of AEAS was found between the two groups. Conclusion The amplification of the BDNF gene influenced neither the therapeutic responses to lithium nor the occurrence of AEAS. Five of 50 patients with bipolar disorders exhibited three copies of the genomic region within exon IV of the BDNF gene. But, 50 healthy subjects had two copies. This amplification did not affect the therapeutic responses to lithium.![]()
Collapse
Affiliation(s)
- Yosuke Suga
- Department of Neuropsychiatry, Kochi Medical School, Kochi University, Nankoku, Japan
| | | | - Shusuke Numata
- Department of Psychiatry, Course of Integrated Brain Sciences, Medical Informatics, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | | | | | - Naoto Kamimura
- Department of Neuropsychiatry, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Ken Sawada
- Department of Neuropsychiatry, Kochi Medical School, Kochi University, Nankoku, Japan.,KOKORONO Support Center, Kochi Health Sciences Center, Ike, Japan
| | - Hiromitsu Kazui
- Department of Neuropsychiatry, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Tetsuro Ohmori
- Department of Psychiatry, Course of Integrated Brain Sciences, Medical Informatics, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Shigeru Morinobu
- Department of Neuropsychiatry, Kochi Medical School, Kochi University, Nankoku, Japan.,Department of Occupational Therapy, School of Health Science and Social Welfare, KIBI International University, Takahashi, Japan
| |
Collapse
|