1
|
Wang Z, Zhang L, Yang J, Zeng Y, Su C, Yao M, Zhang H, Hu W, Liu Y, Lai Y, Wang X, Zeng J, Liu R. Chronic stress induces Alzheimer's disease-like pathologies through DNA damage-Chk1-CIP2A signaling. Aging (Albany NY) 2024; 16:9168-9187. [PMID: 38819231 PMCID: PMC11164505 DOI: 10.18632/aging.205862] [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: 12/19/2023] [Accepted: 03/19/2024] [Indexed: 06/01/2024]
Abstract
Stress is an important initiating factor in promoting Alzheimer's disease (AD) pathogenesis. However, the mechanism by which stress induces AD-like cognitive impairment remains to be clarified. Here, we demonstrate that DNA damage is increased in stress hormone Corticotropin-releasing factor (CRF)-treated cells and in brains of mice exposed to chronic restraint stress. Accumulation of DNA damage drives activation of cell cycle checkpoint protein kinase 1 (Chk1), upregulation of cancerous inhibitor of PP2A (CIP2A), tau hyperphosphorylation, and Aβ overproduction, eventually resulting in synaptic impairment and cognitive deficits. Pharmacological intervention targeting Chk1 by specific inhibitor and DNA damage by vitamin C, suppress DNA damage-Chk1-CIP2A signaling pathway in chronic stress animal model, which in turn attenuate AD-like pathologies, synaptic impairments and cognitive deficits. Our study uncovers a novel molecular mechanism of stress-induced AD-like pathologies and provides effective preventive and therapeutic strategies targeting this signaling pathway.
Collapse
Affiliation(s)
- Zhuoqun Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education/Hubei Province for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lun Zhang
- Department of Pathophysiology, Key Laboratory of Ministry of Education/Hubei Province for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Clinical Laboratory, Wuhan Fourth Hospital, Wuhan, China
| | - Jiayu Yang
- Department of Pathophysiology, Key Laboratory of Ministry of Education/Hubei Province for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Zeng
- Department of Pathophysiology, Key Laboratory of Ministry of Education/Hubei Province for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengke Su
- Department of Pathophysiology, Key Laboratory of Ministry of Education/Hubei Province for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengdong Yao
- Department of Pathophysiology, Key Laboratory of Ministry of Education/Hubei Province for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiliang Zhang
- Department of Pathophysiology, Key Laboratory of Ministry of Education/Hubei Province for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenting Hu
- Department of Pathophysiology, Key Laboratory of Ministry of Education/Hubei Province for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yi Liu
- Department of Pathophysiology, Key Laboratory of Ministry of Education/Hubei Province for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiwen Lai
- Department of Pathophysiology, Key Laboratory of Ministry of Education/Hubei Province for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaochuan Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education/Hubei Province for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, China
| | - Ji Zeng
- Department of Clinical Laboratory, Wuhan Fourth Hospital, Wuhan, China
| | - Rong Liu
- Department of Pathophysiology, Key Laboratory of Ministry of Education/Hubei Province for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, China
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
2
|
Öztürk M, Özkan Y, Yalın Sapmaz Ş, Erdal S, Taneli F, Kandemir H. Thiol/Disulfide Homeostasis: A Potential New Peripheral Biomarker in Adolescent Depression. PSYCHIAT CLIN PSYCH 2024; 34:29-37. [PMID: 38883887 PMCID: PMC11177650 DOI: 10.5152/pcp.2024.23744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 12/10/2023] [Indexed: 06/18/2024] Open
Abstract
Background Thiol-disulfide homeostasis (TDH), one of the most important antioxidants, is involved in the non-enzymatic removal of reactive oxygen molecules in the body and is one of the many methods to measure the level of oxidative stress (OS). In the present study, TDH is investigated in adolescent depression, and its relationship to clinical variables is examined. Methods Thirty-two (50.0%) patients diagnosed with major depressive disorder (MDD) and without psychotropic drug use and 32 (50.0%) healthy controls were included in the present study. The subjects MDD and control groups were between 13 and 18 years old. Participants completed the DSM-5 Level-2 scales for depression and irritability. A colorimetric method proposed by Erel and Neselioglu was used to analyze the TDH parameters of serum samples. Results Biochemical analyses of samples from the MDD and control groups showed significant differences between the groups in native thiol (SH) levels (P = .002), disulfide (SS) levels (P = .021), disulfide/total thiol (SS/ToSH) (P = .009), and disulfide/native thiol (SS/SH) (P = .003) levels. Analysis of receiver operating characteristic showed that the area under the curve values with "acceptable discrimination potential" for the TDH parameters were significantly able to discriminate individuals with MDD from healthy controls. Conclusion Thiol-disulfide homeostasis, one of the OS parameters, was found to be impaired in adolescents with depression. Our results suggest that TDH may contribute to the etiopathogenesis of adolescent MDD and that TDH may be a novel approach to assess OS in adolescent depression.
Collapse
Affiliation(s)
- Masum Öztürk
- Department of Child and Adolescent Psychiatry, Dicle University, Faculty of Medicine, Diyarbakır, Turkey
| | - Yekta Özkan
- Department of Child and Adolescent Psychiatry, Afyonkarahisar University of Health Sciences, Faculty of Medicine, Afyonkarahisar, Turkey
| | - Şermin Yalın Sapmaz
- Department of Child and Adolescent Psychiatry, Manisa Celal Bayar University, Manisa, Turkey
| | - Serkan Erdal
- Department of Biochemistry, Manisa Celal Bayar University, Faculty of Medicine, Manisa, Turkey
| | - Fatma Taneli
- Department of Biochemistry, Manisa Celal Bayar University, Faculty of Medicine, Manisa, Turkey
| | - Hasan Kandemir
- Department of Child and Adolescent Psychiatry, Manisa Celal Bayar University, Manisa, Turkey
| |
Collapse
|
3
|
Abraham M, Peterburs J, Mundorf A. Oligodendrocytes matter: a review of animal studies on early adversity. J Neural Transm (Vienna) 2023; 130:1177-1185. [PMID: 37138023 PMCID: PMC10460720 DOI: 10.1007/s00702-023-02643-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 04/26/2023] [Indexed: 05/05/2023]
Abstract
Exposure to adversities in early life appears to affect the development of white matter, especially oligodendrocytes. Furthermore, altered myelination is present in regions subjected to maturation during the developmental time when early adversities are experienced. In this review, studies applying two well-established animal models of early life adversity, namely maternal separation and maternal immune activation, focusing on oligodendrocyte alterations and resulting implications for psychiatric disorders are discussed. Studies revealed that myelination is reduced as a result of altered oligodendrocyte expression. Furthermore, early adversity is associated with increased cell death, a simpler morphology, and inhibited oligodendrocyte maturation. However, these effects seem to be region- specific as some brain regions show increased expression while others show decreased expression of oligodendroglia-related genes, and they occur especially in regions of ongoing development. Some studies furthermore suggest that early adversity leads to premature differentiation of oligodendrocytes. Importantly, especially early exposure results in stronger oligodendrocyte-related impairments. However, resulting alterations are not restricted to exposure during the early pre- and postnatal days as social isolation after weaning leads to fewer internodes and branches and shorter processes of oligodendrocytes in adulthood. Eventually, the found alterations may lead to dysfunction and long-lasting alterations in structural brain development associated with psychiatric disorders. To date, only few preclinical studies have focused on the effects of early adversity on oligodendrocytes. More studies including several developmental stages are needed to further disentangle the role of oligodendrocytes in the development of psychiatric disorders.
Collapse
Affiliation(s)
- Mate Abraham
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Jutta Peterburs
- Institute for Systems Medicine and Department of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Annakarina Mundorf
- Institute for Systems Medicine and Department of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany.
| |
Collapse
|
4
|
Guo W, Yao X, Cui R, Yang W, Wang L. Mechanisms of paeoniaceae action as an antidepressant. Front Pharmacol 2023; 13:934199. [PMID: 36844911 PMCID: PMC9944447 DOI: 10.3389/fphar.2022.934199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 08/05/2022] [Indexed: 02/10/2023] Open
Abstract
Paeoniflorin (PF) has been widely used for the treatment of depression in mice models, some Chinese herbal compound containing PF on treating depression, such as Xiaoyao San, Chaihu-Shugan-San, Danggui Shaoyao San etc. Many experiments are also verifying whether PF in these powders can be used as an effective component in the treatment of depression. Therefore, in this review the antidepressant effect of PF and its mechanism of action are outlined with particular focus on the following aspects: increasing the levels of monoamine neurotransmitters, inhibiting the HPA axis, promoting neuroprotection, enhancing neurogenesis in the hippocampus, and elevating levels of brain-derived neurotrophic factor (BDNF). This review may be helpful for the application of PF in the treatment of depression.
Collapse
Affiliation(s)
- Wanxu Guo
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Xiaoxiao Yao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Wei Yang
- *Correspondence: Wei Yang, ; Lei Wang,
| | - Lei Wang
- *Correspondence: Wei Yang, ; Lei Wang,
| |
Collapse
|
5
|
Jorgensen A, Baago IB, Rygner Z, Jorgensen MB, Andersen PK, Kessing LV, Poulsen HE. Association of Oxidative Stress-Induced Nucleic Acid Damage With Psychiatric Disorders in Adults: A Systematic Review and Meta-analysis. JAMA Psychiatry 2022; 79:920-931. [PMID: 35921094 PMCID: PMC9350850 DOI: 10.1001/jamapsychiatry.2022.2066] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
IMPORTANCE Nucleic acid damage from oxidative stress (NA-OXS) may be a molecular mechanism driving the severely increased morbidity and mortality from somatic causes in adults with psychiatric disorders. OBJECTIVE To systematically retrieve and analyze data on NA-OXS across the psychiatric disorder diagnostic spectrum. DATA SOURCES The PubMed, Embase, and PsycINFO databases were searched from inception to November 16, 2021. A hand search of reference lists of relevant articles was also performed. STUDY SELECTION Key study inclusion criteria in this meta-analysis were as follows: adult human study population, measurement of any marker of DNA or RNA damage from oxidative stress, and either a (1) cross-sectional design comparing patients with psychiatric disorders (any diagnosis) with a control group or (2) prospective intervention. Two authors screened the studies, and 2 senior authors read the relevant articles in full and assessed them for eligibility. DATA EXTRACTION AND SYNTHESIS The Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines were followed. Two authors performed data extraction independently, and a senior coauthor was consulted in cases of disagreement. Data were synthesized with random-effects and multilevel meta-analyses. MAIN OUTCOMES AND MEASURES The predefined hypothesis was that individuals with psychiatric disorders have increased NA-OXS levels. The main outcome was the standardized mean differences (SMDs) among patients and controls in nucleic acid oxidation markers compared across diagnostic groups. Analyses were divided into combinations of biological matrices and nucleic acids. RESULTS Eighty-two studies fulfilled the inclusion criteria, comprising 205 patient vs control group comparisons and a total of 10 151 patient and 10 532 control observations. Overall, the data showed that patients with psychiatric disorders had higher NA-OXS levels vs controls across matrices and molecules. Pooled effect sizes ranged from moderate for urinary DNA markers (SMD = 0.44 [95% CI, 0.20-0.68]; P < .001) to very large for blood cell DNA markers (SMD = 1.12 [95% CI, 0.69-1.55; P < .001). Higher NA-OXS levels were observed among patients with dementias followed by psychotic and bipolar disorders. Sensitivity analyses excluding low-quality studies did not materially alter the results. Intervention studies were few and too heterogenous for meaningful meta-analysis. CONCLUSIONS AND RELEVANCE The results of this meta-analysis suggest that there is an association with increased NA-OXS levels in individuals across the psychiatric disorder diagnostic spectrum. NA-OXS may play a role in the somatic morbidity and mortality observed among individuals with psychiatric disorders.
Collapse
Affiliation(s)
- Anders Jorgensen
- Psychiatric Center Copenhagen, Mental Health Services Copenhagen, Copenhagen, Denmark,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ida Bendixen Baago
- Psychiatric Center Copenhagen, Mental Health Services Copenhagen, Copenhagen, Denmark
| | - Zerlina Rygner
- Psychiatric Center Copenhagen, Mental Health Services Copenhagen, Copenhagen, Denmark,Department of Cardiology, Copenhagen University Hospital, Hillerød, Denmark,Department of Endocrinology, Copenhagen University Hospital, Hillerød, Denmark
| | - Martin Balslev Jorgensen
- Psychiatric Center Copenhagen, Mental Health Services Copenhagen, Copenhagen, Denmark,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | - Lars Vedel Kessing
- Psychiatric Center Copenhagen, Mental Health Services Copenhagen, Copenhagen, Denmark,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Enghusen Poulsen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark,Department of Cardiology, Copenhagen University Hospital, Hillerød, Denmark,Department of Endocrinology, Copenhagen University Hospital, Hillerød, Denmark
| |
Collapse
|
6
|
Abraham M, Mundorf A, Brodmann K, Freund N. Unraveling the mystery of white matter in depression: A translational perspective on recent advances. Brain Behav 2022; 12:e2629. [PMID: 35652161 PMCID: PMC9304855 DOI: 10.1002/brb3.2629] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/15/2022] [Accepted: 04/23/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Numerous cortical and subcortical structures have been studied extensively concerning alterations of their integrity as well as their neurotransmitters in depression. However, connections between these structures have received considerably less attention. OBJECTIVE This systematic review presents results from recent neuroimaging as well as neuropathologic studies conducted on humans and other mammals. It aims to provide evidence for impaired white matter integrity in individuals expressing a depressive phenotype. METHODS A systematic database search in accordance with the PRISMA guidelines was conducted to identify imaging and postmortem studies conducted on humans with a diagnosis of major depressive disorder, as well as on rodents and primates subjected to an animal model of depression. RESULTS Alterations are especially apparent in frontal gyri, as well as in structures establishing interhemispheric connectivity between frontal regions. Translational neuropathological findings point to alterations in oligodendrocyte density and morphology, as well as to alterations in the expression of genes related to myelin synthesis. An important role of early life adversities in the development of depressive symptoms and white matter alterations across species is thereby revealed. Data indicating that stress can interfere with physiological myelination patterns is presented. Altered myelination is most notably present in regions that are subject to maturation during the developmental stage of exposure to adversities. CONCLUSION Translational studies point to replicable alterations in white matter integrity in subjects suffering from depression across multiple species. Impaired white matter integrity is apparent in imaging as well as neuropathological studies. Future studies should focus on determining to what extent influencing white matter integrity is able to improve symptoms of depression in animals as well as humans.
Collapse
Affiliation(s)
- Mate Abraham
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Annakarina Mundorf
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University Bochum, Bochum, Germany.,Institute for Systems Medicine and Department of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Katja Brodmann
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Nadja Freund
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
7
|
Riveros ME, Ávila A, Schruers K, Ezquer F. Antioxidant Biomolecules and Their Potential for the Treatment of Difficult-to-Treat Depression and Conventional Treatment-Resistant Depression. Antioxidants (Basel) 2022; 11:540. [PMID: 35326190 PMCID: PMC8944633 DOI: 10.3390/antiox11030540] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 11/23/2022] Open
Abstract
Major depression is a devastating disease affecting an increasing number of people from a young age worldwide, a situation that is expected to be worsened by the COVID-19 pandemic. New approaches for the treatment of this disease are urgently needed since available treatments are not effective for all patients, take a long time to produce an effect, and are not well-tolerated in many cases; moreover, they are not safe for all patients. There is solid evidence showing that the antioxidant capacity is lower and the oxidative damage is higher in the brains of depressed patients as compared with healthy controls. Mitochondrial disfunction is associated with depression and other neuropsychiatric disorders, and this dysfunction can be an important source of oxidative damage. Additionally, neuroinflammation that is commonly present in the brain of depressive patients highly contributes to the generation of reactive oxygen species (ROS). There is evidence showing that pro-inflammatory diets can increase depression risk; on the contrary, an anti-inflammatory diet such as the Mediterranean diet can decrease it. Therefore, it is interesting to evaluate the possible role of plant-derived antioxidants in depression treatment and prevention as well as other biomolecules with high antioxidant and anti-inflammatory potential such as the molecules paracrinely secreted by mesenchymal stem cells. In this review, we evaluated the preclinical and clinical evidence showing the potential effects of different antioxidant and anti-inflammatory biomolecules as antidepressants, with a focus on difficult-to-treat depression and conventional treatment-resistant depression.
Collapse
Affiliation(s)
- María Eugenia Riveros
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Santiago 7710162, Chile
| | - Alba Ávila
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Santiago 7710162, Chile;
| | - Koen Schruers
- Department of Psychiatry and Neuropsychology, Maastricht University Medical Center, 6229 Maastricht, The Netherlands;
| | - Fernando Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Santiago 7710162, Chile;
| |
Collapse
|
8
|
Wang Y, Pleasure D, Deng W, Guo F. Therapeutic Potentials of Poly (ADP-Ribose) Polymerase 1 (PARP1) Inhibition in Multiple Sclerosis and Animal Models: Concept Revisiting. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2102853. [PMID: 34935305 PMCID: PMC8844485 DOI: 10.1002/advs.202102853] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 10/12/2021] [Indexed: 05/05/2023]
Abstract
Poly (ADP-ribose) polymerase 1 (PARP1) plays a fundamental role in DNA repair and gene expression. Excessive PARP1 hyperactivation, however, has been associated with cell death. PARP1 and/or its activity are dysregulated in the immune and central nervous system of multiple sclerosis (MS) patients and animal models. Pharmacological PARP1 inhibition is shown to be protective against immune activation and disease severity in MS animal models while genetic PARP1 deficiency studies reported discrepant results. The inconsistency suggests that the function of PARP1 and PARP1-mediated PARylation may be complex and context-dependent. The article reviews PARP1 functions, discusses experimental findings and possible interpretations of PARP1 in inflammation, neuronal/axonal degeneration, and oligodendrogliopathy, three major pathological components cooperatively determining MS disease course and neurological progression, and points out future research directions. Cell type specific PARP1 manipulations are necessary for revisiting the role of PARP1 in the three pathological components prior to moving PARP1 inhibition into clinical trials for MS therapy.
Collapse
Affiliation(s)
- Yan Wang
- Department of NeurologySchool of MedicineUniversity of CaliforniaDavisCA95817USA
- Institute for Pediatric Regenerative MedicineUC Davis School of Medicine/Shriners Hospitals for ChildrenSacramentoCAUSA
| | - David Pleasure
- Department of NeurologySchool of MedicineUniversity of CaliforniaDavisCA95817USA
- Institute for Pediatric Regenerative MedicineUC Davis School of Medicine/Shriners Hospitals for ChildrenSacramentoCAUSA
| | - Wenbin Deng
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityGuangzhou510006China
| | - Fuzheng Guo
- Department of NeurologySchool of MedicineUniversity of CaliforniaDavisCA95817USA
- Institute for Pediatric Regenerative MedicineUC Davis School of Medicine/Shriners Hospitals for ChildrenSacramentoCAUSA
| |
Collapse
|
9
|
Nelson MM, Hoff JD, Zeese ML, Corfas G. Poly (ADP-Ribose) Polymerase 1 Regulates Cajal-Retzius Cell Development and Neural Precursor Cell Adhesion. Front Cell Dev Biol 2021; 9:693595. [PMID: 34708032 PMCID: PMC8542860 DOI: 10.3389/fcell.2021.693595] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 09/16/2021] [Indexed: 11/13/2022] Open
Abstract
Poly (ADP-ribose) polymerase 1 (PARP1) is a ubiquitously expressed enzyme that regulates DNA damage repair, cell death, inflammation, and transcription. PARP1 functions by adding ADP-ribose polymers (PAR) to proteins including itself, using NAD+ as a donor. This post-translational modification known as PARylation results in changes in the activity of PARP1 and its substrate proteins and has been linked to the pathogenesis of various neurological diseases. PARP1 KO mice display schizophrenia-like behaviors, have impaired memory formation, and have defects in neuronal proliferation and survival, while mutations in genes that affect PARylation have been associated with intellectual disability, psychosis, neurodegeneration, and stroke in humans. Yet, the roles of PARP1 in brain development have not been extensively studied. We now find that loss of PARP1 leads to defects in brain development and increased neuronal density at birth. We further demonstrate that PARP1 loss increases the expression levels of genes associated with neuronal migration and adhesion in the E15.5 cerebral cortex, including Reln. This correlates with an increased number of Cajal–Retzius (CR) cells in vivo and in cultures of embryonic neural progenitor cells (NPCs) derived from the PARP1 KO cortex. Furthermore, PARP1 loss leads to increased NPC adhesion to N-cadherin, like that induced by experimental exposure to Reelin. Taken together, these results uncover a novel role for PARP1 in brain development, i.e., regulation of CR cells, neuronal density, and cell adhesion.
Collapse
Affiliation(s)
- Megan M Nelson
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, MI, United States.,Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, United States
| | - J Damon Hoff
- Single Molecule Analysis in Real-Time Center, Department of Biophysics, University of Michigan, Ann Arbor, MI, United States
| | - Mya L Zeese
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, MI, United States.,Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Gabriel Corfas
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, MI, United States.,Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
10
|
Lan T, Li Y, Fan C, Wang L, Wang W, Chen S, Yu SY. MicroRNA-204-5p reduction in rat hippocampus contributes to stress-induced pathology via targeting RGS12 signaling pathway. J Neuroinflammation 2021; 18:243. [PMID: 34674723 PMCID: PMC8532383 DOI: 10.1186/s12974-021-02299-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/17/2021] [Indexed: 11/18/2022] Open
Abstract
Background Neuroinflammation occupies a pivotal position in the pathogenesis of most nervous system diseases, including depression. However, the underlying molecular mechanisms of neuroinflammation associated with neuronal injury in depression remain largely uncharacterized. Therefore, identifying potential molecular mechanisms and therapeutic targets would serve to better understand the progression of this condition. Methods Chronic unpredictable stress (CUS) was used to induce depression-like behaviors in rats. RNA-sequencing was used to detect the differentially expressed microRNAs. Stereotactic injection of AAV virus to overexpress or knockdown the miR-204-5p. The oxidative markers and inflammatory related proteins were verified by immunoblotting or immunofluorescence assay. The oxidative stress enzyme and products were verified using enzyme-linked assay kit. Electron microscopy analysis was used to observe the synapse and ultrastructural pathology. Finally, electrophysiological recording was used to analyze the synaptic transmission. Results Here, we found that the expression of miR-204-5p within the hippocampal dentate gyrus (DG) region of rats was significantly down-regulated after chronic unpredicted stress (CUS), accompanied with the oxidative stress-induced neuronal damage within DG region of these rats. In contrast, overexpression of miR-204-5p within the DG region of CUS rats alleviated oxidative stress and neuroinflammation by directly targeting the regulator of G protein signaling 12 (RGS12), effects which were accompanied with amelioration of depressive-like behaviors in these CUS rats. In addition, down-regulation of miR-204-5p induced neuronal deterioration in DG regions and depressive-like behaviors in rats. Conclusion Taken together, these results suggest that miR-204-5p plays a key role in regulating oxidative stress damage in CUS-induced pathological processes of depression. Such findings provide evidence of the involvement of miR-204-5p in mechanisms underlying oxidative stress associated with depressive phenotype. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02299-5.
Collapse
Affiliation(s)
- Tian Lan
- Department of Physiology, School of Basic Medical Sciences, Shandong University, 44 Wenhuaxilu Road, Jinan, Shandong, 250012, People's Republic of China
| | - Ye Li
- Department of Physiology, School of Basic Medical Sciences, Shandong University, 44 Wenhuaxilu Road, Jinan, Shandong, 250012, People's Republic of China
| | - Cuiqin Fan
- Department of Physiology, School of Basic Medical Sciences, Shandong University, 44 Wenhuaxilu Road, Jinan, Shandong, 250012, People's Republic of China
| | - Liyan Wang
- Morphological Experimental Center, School of Basic Medical Sciences, Shandong University, 44 Wenhuaxilu Road, Jinan, Shandong, 250012, People's Republic of China
| | - Wenjing Wang
- Department of Physiology, School of Basic Medical Sciences, Shandong University, 44 Wenhuaxilu Road, Jinan, Shandong, 250012, People's Republic of China
| | - Shihong Chen
- Department of Endocrinology, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong, 250033, People's Republic of China.
| | - Shu Yan Yu
- Department of Physiology, School of Basic Medical Sciences, Shandong University, 44 Wenhuaxilu Road, Jinan, Shandong, 250012, People's Republic of China. .,Shandong Provincial Key Laboratory of Mental Disorders, School of Basic Medical Sciences, 44 Wenhuaxilu Road, Jinan, Shandong, 250012, People's Republic of China.
| |
Collapse
|
11
|
Schossler Garcia C, Garcia PR, da Silva Espíndola CN, Nunes GD, Jardim NS, Müller SG, Bortolatto CF, Brüning CA. Effect of m-Trifluoromethyl-diphenyl diselenide on the Pain-Depression Dyad Induced by Reserpine: Insights on Oxidative Stress, Apoptotic, and Glucocorticoid Receptor Modulation. Mol Neurobiol 2021; 58:5078-5089. [PMID: 34245440 DOI: 10.1007/s12035-021-02483-x] [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: 02/19/2021] [Accepted: 07/02/2021] [Indexed: 01/07/2023]
Abstract
Chronic pain and depression often coexist sharing common pathological mechanisms, and available analgesics and antidepressants have demonstrated limited clinical efficacy. Evidence has demonstrated that neuronal oxidative stress, apoptosis, and also glucocorticoid receptor dysregulation facilitate the occurrence and development of both chronic pain and depression. This study evaluated the effect of the organoselenium compound m-trifluoromethyl-diphenyl diselenide [(m-CF3-PhSe)2] in the pain-depression comorbidity induced by reserpine. Mice were treated with reserpine 0.5 mg/kg for 3 days (intraperitoneal, once a day), and in the next 2 days, they were treated with (m-CF3-PhSe)2 10 mg/kg (intragastric, once a day). Thirty minutes after the last administration of (m-CF3-PhSe)2, mice were subjected to the behavioral testing. (m-CF3-PhSe)2 treatment reverted the reserpine-increased thermal hyperalgesia and depressive-like behavior observed in the hot-plate test and forced swimming test, respectively. Reserpine provoked a decrease of crossings and rearings in the open-field test, while (m-CF3-PhSe)2 presented a tendency to normalize these parameters. Reserpine and/or (m-CF3-PhSe)2 treatments did not alter the locomotor activity of mice observed in the rota-rod test. These effects could be related to modulation of oxidative stress, apoptotic pathway, and glucocorticoid receptors, once (m-CF3-PhSe)2 normalized thiobarbituric acid reactive substances and 4-hydroxynonenal modified protein levels, markers of lipoperoxidation, poly(ADP-ribose) polymerase cleaved/total ratio, and glucocorticoid receptor levels increased by reserpine in the hippocampus. Considering that pain-depression dyad is a complex state of difficult treatment, this organoselenium compound could raise as an interesting alternative to treat pain-depression condition.
Collapse
Affiliation(s)
- Cleisson Schossler Garcia
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center for Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Pelotas, RS, 96010-900, Brazil
| | - Pabliane Rodrigues Garcia
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center for Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Pelotas, RS, 96010-900, Brazil
| | - Carlos Natã da Silva Espíndola
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center for Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Pelotas, RS, 96010-900, Brazil
| | - Gustavo D'Avila Nunes
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center for Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Pelotas, RS, 96010-900, Brazil
| | - Natália Silva Jardim
- Laboratory of Synthesis, Reactivity and Pharmacological and Toxicological Evaluation of Organochalcogens, Federal University of Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | - Sabrina Grendene Müller
- Laboratory of Synthesis, Reactivity and Pharmacological and Toxicological Evaluation of Organochalcogens, Federal University of Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | - Cristiani Folharini Bortolatto
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center for Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Pelotas, RS, 96010-900, Brazil.
| | - César Augusto Brüning
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center for Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Pelotas, RS, 96010-900, Brazil.
| |
Collapse
|
12
|
Szopa A, Bogatko K, Herbet M, Serefko A, Ostrowska M, Wośko S, Świąder K, Szewczyk B, Wlaź A, Skałecki P, Wróbel A, Mandziuk S, Pochodyła A, Kudela A, Dudka J, Radziwoń-Zaleska M, Wlaź P, Poleszak E. The Interaction of Selective A1 and A2A Adenosine Receptor Antagonists with Magnesium and Zinc Ions in Mice: Behavioural, Biochemical and Molecular Studies. Int J Mol Sci 2021; 22:ijms22041840. [PMID: 33673282 PMCID: PMC7918707 DOI: 10.3390/ijms22041840] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 02/06/2023] Open
Abstract
The purpose of the study was to investigate whether the co-administration of Mg2+ and Zn2+ with selective A1 and A2A receptor antagonists might be an interesting antidepressant strategy. Forced swim, tail suspension, and spontaneous locomotor motility tests in mice were performed. Further, biochemical and molecular studies were conducted. The obtained results indicate the interaction of DPCPX and istradefylline with Mg2+ and Zn2+ manifested in an antidepressant-like effect. The reduction of the BDNF serum level after co-administration of DPCPX and istradefylline with Mg2+ and Zn2+ was noted. Additionally, Mg2+ or Zn2+, both alone and in combination with DPCPX or istradefylline, causes changes in Adora1 expression, DPCPX or istradefylline co-administered with Zn2+ increases Slc6a15 expression as compared to a single-drug treatment, co-administration of tested agents does not have a more favourable effect on Comt expression. Moreover, the changes obtained in Ogg1, MsrA, Nrf2 expression show that DPCPX-Mg2+, DPCPX-Zn2+, istradefylline-Mg2+ and istradefylline-Zn2+ co-treatment may have greater antioxidant capacity benefits than administration of DPCPX and istradefylline alone. It seems plausible that a combination of selective A1 as well as an A2A receptor antagonist and magnesium or zinc may be a new antidepressant therapeutic strategy.
Collapse
Affiliation(s)
- Aleksandra Szopa
- Chair and Department of Applied and Social Pharmacy, Laboratory of Preclinical Testing, Medical University of Lublin, 1 Chodźki Street, PL 20–093 Lublin, Poland; (K.B.); (A.S.); (S.W.)
- Correspondence: (A.S.); (E.P.)
| | - Karolina Bogatko
- Chair and Department of Applied and Social Pharmacy, Laboratory of Preclinical Testing, Medical University of Lublin, 1 Chodźki Street, PL 20–093 Lublin, Poland; (K.B.); (A.S.); (S.W.)
| | - Mariola Herbet
- Chair and Department of Toxicology, Medical University of Lublin, 8 Chodźki Street, PL 20–093 Lublin, Poland; (M.H.); (M.O.); (A.K.) (J.D.)
| | - Anna Serefko
- Chair and Department of Applied and Social Pharmacy, Laboratory of Preclinical Testing, Medical University of Lublin, 1 Chodźki Street, PL 20–093 Lublin, Poland; (K.B.); (A.S.); (S.W.)
| | - Marta Ostrowska
- Chair and Department of Toxicology, Medical University of Lublin, 8 Chodźki Street, PL 20–093 Lublin, Poland; (M.H.); (M.O.); (A.K.) (J.D.)
| | - Sylwia Wośko
- Chair and Department of Applied and Social Pharmacy, Laboratory of Preclinical Testing, Medical University of Lublin, 1 Chodźki Street, PL 20–093 Lublin, Poland; (K.B.); (A.S.); (S.W.)
| | - Katarzyna Świąder
- Chair and Department of Applied and Social Pharmacy, Medical University of Lublin, 1 Chodźki Street, PL 20–093 Lublin, Poland; (K.Ś.); (A.P.)
| | - Bernadeta Szewczyk
- Department of Neurobiology, Polish Academy of Sciences, Maj Institute of Pharmacology, 12 Smętna Street, PL 31–343 Kraków, Poland;
| | - Aleksandra Wlaź
- Department of Pathophysiology, Medical University of Lublin, 8 Jaczewskiego Street, PL 20–090 Lublin, Poland;
| | - Piotr Skałecki
- Department of Commodity Science and Processing of Raw Animal Materials, University of Life Sciences, 13 Akademicka Street, PL 20–950 Lublin, Poland;
| | - Andrzej Wróbel
- Second Department of Gynecology, 8 Jaczewskiego Street, PL 20–090 Lublin, Poland;
| | - Sławomir Mandziuk
- Department of Pneumology, Oncology and Allergology, Medical University of Lublin, 8 Jaczewskiego Street, PL 20–090 Lublin, Poland;
| | - Aleksandra Pochodyła
- Chair and Department of Applied and Social Pharmacy, Medical University of Lublin, 1 Chodźki Street, PL 20–093 Lublin, Poland; (K.Ś.); (A.P.)
| | - Anna Kudela
- Chair and Department of Toxicology, Medical University of Lublin, 8 Chodźki Street, PL 20–093 Lublin, Poland; (M.H.); (M.O.); (A.K.) (J.D.)
| | - Jarosław Dudka
- Chair and Department of Toxicology, Medical University of Lublin, 8 Chodźki Street, PL 20–093 Lublin, Poland; (M.H.); (M.O.); (A.K.) (J.D.)
| | - Maria Radziwoń-Zaleska
- Department of Psychiatry, Medical University of Warsaw, 27 Nowowiejska Street, PL 00–665 Warsaw, Poland;
| | - Piotr Wlaź
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie–Skłodowska University, Akademicka 19, PL 20–033 Lublin, Poland;
| | - Ewa Poleszak
- Chair and Department of Applied and Social Pharmacy, Laboratory of Preclinical Testing, Medical University of Lublin, 1 Chodźki Street, PL 20–093 Lublin, Poland; (K.B.); (A.S.); (S.W.)
- Correspondence: (A.S.); (E.P.)
| |
Collapse
|
13
|
Jewett BE, Miller MN, Ligon LA, Carter Z, Mohammad I, Ordway GA. Rapid and temporary improvement of depression and anxiety observed following niraparib administration: a case report. BMC Psychiatry 2020; 20:171. [PMID: 32295563 PMCID: PMC7161116 DOI: 10.1186/s12888-020-02590-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/07/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Cancer patients are disproportionately affected by generalized anxiety and major depression. For many, current treatments for these conditions are ineffective. In this case report, we present a serendipitous case of anxiety and depression improvement following administration of the poly (ADP-ribose) polymerase (PARP) inhibitor niraparib. CASE PRESENTATION A 61-year old woman with a 20-year history of mild depression developed recurrent ovarian carcinoma and was placed on niraparib for maintenance chemotherapy. With the original onset of ovarian cancer, she experienced an episode of major depression that was resolved with sertraline. After recurrence of ovarian cancer, she experienced a recurrence of major depression and a new onset of generalized anxiety that failed to completely respond to multiple medications. After beginning niraparib therapy the patient noticed a rapid resolution of the symptoms of her anxiety and depression, an effect that was limited to 10-14 days. Due to bone marrow suppression, the patient was taken off and restarted on niraparib several times. Each discontinuation of niraparib resulted in return of her depression and anxiety, while each recontinuation of niraparib resulted in an improvement in her mood and anxiety. CONCLUSIONS This case demonstrates rapid and temporary improvement of anxiety and depression following niraparib administration. There is ample preclinical data that PARP signaling may play a role in psychiatric illness. A small amount of indirect data from clinical trials also shows that niraparib may have psychiatric benefits. Further research on PARP inhibition and its potential psychoactive effects is sorely needed.
Collapse
Affiliation(s)
- Benjamin E. Jewett
- Department of Biomedical Sciences, PO Box 70577, Johnson City, 37614 USA
| | - Merry N. Miller
- grid.255381.80000 0001 2180 1673Department of Psychiatry and Behavioral Sciences, East Tennessee State University, 187 Maple Avenue, Johnson City, TN 37684 USA
| | - Libby A. Ligon
- Department of Biomedical Sciences, PO Box 70577, Johnson City, 37614 USA
| | - Zachary Carter
- Department of Biomedical Sciences, PO Box 70577, Johnson City, 37614 USA
| | - Ibrahim Mohammad
- Department of Biomedical Sciences, PO Box 70577, Johnson City, 37614 USA
| | - Gregory A. Ordway
- Department of Biomedical Sciences, PO Box 70577, Johnson City, 37614 USA ,grid.255381.80000 0001 2180 1673Department of Psychiatry and Behavioral Sciences, East Tennessee State University, 187 Maple Avenue, Johnson City, TN 37684 USA
| |
Collapse
|
14
|
Chen WW, Fu WY, Su YT, Fang WQ, Fu AKY, Ip NY. Increased Axin expression enhances adult hippocampal neurogenesis and exerts an antidepressant effect. Sci Rep 2019; 9:1190. [PMID: 30718786 PMCID: PMC6362220 DOI: 10.1038/s41598-018-38103-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 12/18/2018] [Indexed: 01/21/2023] Open
Abstract
Major depressive disorders are emerging health problems that affect millions of people worldwide. However, treatment options and targets for drug development are limited. Impaired adult hippocampal neurogenesis is emerging as a key contributor to the pathology of major depressive disorders. We previously demonstrated that increasing the expression of the multifunctional scaffold protein Axis inhibition protein (Axin) by administration of the small molecule XAV939 enhances embryonic neurogenesis and affects social interaction behaviors. This prompted us to examine whether increasing Axin protein level can enhance adult hippocampal neurogenesis and thus contribute to mood regulation. Here, we report that stabilizing Axin increases adult hippocampal neurogenesis and exerts an antidepressant effect. Specifically, treating adult mice with XAV939 increased the amplification of adult neural progenitor cells and neuron production in the hippocampus under both normal and chronic stress conditions. Furthermore, XAV939 injection in mice ameliorated depression-like behaviors induced by chronic restraint stress. Thus, our study demonstrates that Axin/XAV939 plays an important role in adult hippocampal neurogenesis and provides a potential therapeutic approach for mood-related disorders.
Collapse
Affiliation(s)
- Wei-Wei Chen
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Wing-Yu Fu
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Yi-Ting Su
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Wei-Qun Fang
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Amy K Y Fu
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, Guangdong, China
| | - Nancy Y Ip
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China. .,Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China. .,State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China. .,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, Guangdong, China.
| |
Collapse
|
15
|
Morris G, Walker AJ, Berk M, Maes M, Puri BK. Cell Death Pathways: a Novel Therapeutic Approach for Neuroscientists. Mol Neurobiol 2018; 55:5767-5786. [PMID: 29052145 PMCID: PMC5994217 DOI: 10.1007/s12035-017-0793-y] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 09/26/2017] [Indexed: 02/08/2023]
Abstract
In the first part, the following mechanisms involved in different forms of cell death are considered, with a view to identifying potential therapeutic targets: tumour necrosis factor receptors (TNFRs) and their engagement by tumour necrosis factor-alpha (TNF-α); poly [ADP-ribose] polymerase (PARP)-1 cleavage; the apoptosis signalling kinase (ASK)-c-Jun N-terminal kinase (JNK) axis; lysosomal permeability; activation of programmed necrotic cell death; oxidative stress, caspase-3 inhibition and parthanatos; activation of inflammasomes by reactive oxygen species and the development of pyroptosis; oxidative stress, calcium dyshomeostasis and iron in the development of lysosomal-mediated necrosis and lysosomal membrane permeability; and oxidative stress, lipid peroxidation, iron dyshomeostasis and ferroptosis. In the second part, there is a consideration of the role of lethal and sub-lethal activation of these pathways in the pathogenesis and pathophysiology of neurodegenerative and neuroprogressive disorders, with particular reference to the TNF-α-TNFR signalling axis; dysregulation of ASK-1-JNK signalling; prolonged or chronic PARP-1 activation; the role of pyroptosis and chronic inflammasome activation; and the roles of lysosomal permeabilisation, necroptosis and ferroptosis. Finally, it is suggested that, in addition to targeting oxidative stress and inflammatory processes generally, neuropsychiatric disorders may respond to therapeutic targeting of TNF-α, PARP-1, the Nod-like receptor NLRP3 inflammasome and the necrosomal molecular switch receptor-interacting protein kinase-3, since their widespread activation can drive and/or exacerbate peripheral inflammation and neuroinflammation even in the absence of cell death. To this end, the use is proposed of a combination of the tetracycline derivative minocycline and N-acetylcysteine as adjunctive treatment for a range of neuropsychiatric disorders.
Collapse
Affiliation(s)
- G Morris
- , Bryn Road Seaside 87, Llanelli, Wales, , SA15 2LW, UK
- School of Medicine, Deakin University, Geelong, 3220, Australia
| | - A J Walker
- School of Medicine, Deakin University, Geelong, 3220, Australia
| | - M Berk
- The Centre for Molecular and Medical Research, School of Medicine, Deakin University, P.O. Box 291, Geelong, 3220, Australia
- Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, 60430-040, Brazil
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, P.O. Box 291, Geelong, 3220, Australia
- Orygen Youth Health Research Centre and the Centre of Youth Mental Health, The Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, University of Melbourne, Parkville, 3052, Australia
| | - M Maes
- School of Medicine, Deakin University, Geelong, 3220, Australia
- Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
| | - B K Puri
- Department of Medicine, Hammersmith Hospital, Imperial College London, London, W12 0HS, UK.
| |
Collapse
|
16
|
Astrocyte pathology in the ventral prefrontal white matter in depression. J Psychiatr Res 2018; 102:150-158. [PMID: 29660602 PMCID: PMC6005746 DOI: 10.1016/j.jpsychires.2018.04.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 03/22/2018] [Accepted: 04/05/2018] [Indexed: 12/15/2022]
Abstract
Astrocyte functions in white matter are less well understood than in gray matter. Our recent study of white matter in ventral prefrontal cortex (vPFC) revealed alterations in expression of myelin-related genes in major depressive disorder (MDD). Since white matter astrocytes maintain myelin, we hypothesized that morphometry of these cells will be altered in MDD in the same prefrontal white matter region in which myelin-related genes are altered. White matter adjacent to vPFC was examined in 25 MDD and 21 control subjects. Density and size of GFAP-immunoreactive (-ir) astrocyte cell bodies was measured. The area fraction of GFAP-ir astrocytes (cell bodies + processes) was also estimated. GFAP mRNA expression was determined using qRT-PCR. The density of GFAP-ir astrocytes was also measured in vPFC white matter of rats subjected to chronic unpredictable stress (CUS) and control animals. Fibrous and smooth GFAP-ir astrocytes were distinguished in human white matter. The density of both types of astrocytes was significantly decreased in MDD. Area fraction of GFAP immunoreactivity was significantly decreased in MDD, but mean soma size remained unchanged. Expression of GFAP mRNA was significantly decreased in MDD. In CUS rats there was a significant decrease in astrocyte density in prefrontal white matter. The decrease in density and area fraction of white matter astrocytes and GFAP mRNA in MDD may be linked to myelin pathology previously noted in these subjects. Astrocyte pathology may contribute to axon disturbances in axon integrity reported by neuroimaging studies in MDD and interfere with signal conduction in the white matter.
Collapse
|
17
|
Oxidative stress and DNA damage after cerebral ischemia: Potential therapeutic targets to repair the genome and improve stroke recovery. Neuropharmacology 2017; 134:208-217. [PMID: 29128308 DOI: 10.1016/j.neuropharm.2017.11.011] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/02/2017] [Accepted: 11/05/2017] [Indexed: 12/12/2022]
Abstract
The past two decades have witnessed remarkable advances in oxidative stress research, particularly in the context of ischemic brain injury. Oxidative stress in ischemic tissues compromises the integrity of the genome, resulting in DNA lesions, cell death in neurons, glial cells, and vascular cells, and impairments in neurological recovery after stroke. As DNA is particularly vulnerable to oxidative attack, cells have evolved the ability to induce multiple DNA repair mechanisms, including base excision repair (BER), nucleotide excision repair (NER) and non-homogenous endpoint jointing (NHEJ). Defective DNA repair is tightly correlated with worse neurological outcomes after stroke, whereas upregulation of DNA repair enzymes, such as APE1, OGG1, and XRCC1, improves long-term functional recovery following stroke. Indeed, DNA damage and repair are now known to play critical roles in fundamental aspects of stroke recovery, such as neurogenesis, white matter recovery, and neurovascular unit remodeling. Several DNA repair enzymes are essential for comprehensive neural repair mechanisms after stroke, including Polβ and NEIL3 for neurogenesis, APE1 for white matter repair, Gadd45b for axonal regeneration, and DNA-PKs for neurovascular remodeling. This review discusses the emerging role of DNA damage and repair in functional recovery after stroke and highlights the contribution of DNA repair to regenerative elements after stroke. This article is part of the Special Issue entitled 'Cerebral Ischemia'.
Collapse
|
18
|
Maluach AM, Misquitta KA, Prevot TD, Fee C, Sibille E, Banasr M, Andreazza AC. Increased Neuronal DNA/RNA Oxidation in the Frontal Cortex of Mice Subjected to Unpredictable Chronic Mild Stress. CHRONIC STRESS 2017; 1. [PMID: 29250610 PMCID: PMC5730347 DOI: 10.1177/2470547017724744] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Chronic stress is implicated in the development of various psychiatric
illnesses including major depressive disorder. Previous reports suggest that
patients with major depressive disorder have increased levels of oxidative
stress, including higher levels of DNA/RNA oxidation found in postmortem
studies, especially within brain regions responsible for the cognitive and
emotional processes disrupted in the disorder. Here, we aimed to investigate
whether unpredictable chronic mild stress in mice induces neuronal DNA/RNA
oxidation in the prelimbic, infralimbic, and cingulate cortices of the
frontal cortex and the basolateral amygdala and to explore potential
associations with depressive-like behaviors. We expected that animals
subjected to unpredictable chronic mild stress will present higher levels of
DNA/RNA oxidation, which will be associated with anxiety-/depressive-like
behaviors. Methods C57BL/6J mice were assigned to unpredictable chronic mild stress or nonstress
conditions (n = 10/group, 50% females). Following five weeks of
unpredictable chronic mild stress exposure, mice were tested in a series of
behavioral tests measuring anxiety- and depressive-like behaviors. Frontal
cortex and amygdala sections were then immunolabeled for neuronal nuclei, a
marker of post-mitotic neurons and
anti-8-hydroxy-2-deoxyguanosine/8-oxo-7,8-dihydroguanosine, which reflects
both DNA and RNA oxidation. Results Levels of neuronal DNA/RNA oxidation were increased in the frontal cortex of
mice subjected to unpredictable chronic mild stress (p = 0.0207). Levels of neuronal DNA/RNA oxidation in the
frontal cortex were positively correlated with z-emotionality scores for
latency to feed in the novelty-suppressed feeding test (p = 0.0031). Statistically significant differences were not
detected in basolateral amygdala levels of neuronal DNA/RNA oxidation
between nonstress- and unpredictable chronic mild stress-exposed mice, nor
were correlations found with behavioral performances for this region. Conclusion Our results demonstrate that unpredictable chronic mild stress induces a
significant increase in neuronal DNA/RNA oxidation in the frontal cortex
that correlate with behavioral readouts of the stress response. A lack of
DNA/RNA oxidation alterations in the basolateral amygdala suggests greater
vulnerability of frontal cortex neurons to DNA/RNA oxidation in response to
unpredictable chronic mild stress. These findings add support to the
hypothesis that chronic stress-induced damage to DNA/RNA may be an
additional molecular mechanism underlying cellular dysfunctions associated
with chronic stress and present in stress-related disorders.
Collapse
Affiliation(s)
- Alfred M Maluach
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Keith A Misquitta
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada.,Department of Psychiatry, University of Toronto, Toronto, Canada.,Campbell Family Mental Health Research Institute of CAMH, Toronto, Ontario, Canada
| | - Thomas D Prevot
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada.,Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Corey Fee
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada.,Department of Psychiatry, University of Toronto, Toronto, Canada.,Campbell Family Mental Health Research Institute of CAMH, Toronto, Ontario, Canada
| | - Etienne Sibille
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada.,Department of Psychiatry, University of Toronto, Toronto, Canada.,Campbell Family Mental Health Research Institute of CAMH, Toronto, Ontario, Canada
| | - Mounira Banasr
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada.,Department of Psychiatry, University of Toronto, Toronto, Canada.,Campbell Family Mental Health Research Institute of CAMH, Toronto, Ontario, Canada
| | - Ana C Andreazza
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada.,Department of Psychiatry, University of Toronto, Toronto, Canada.,Campbell Family Mental Health Research Institute of CAMH, Toronto, Ontario, Canada
| |
Collapse
|
19
|
Ordway GA, Szebeni A, Hernandez LJ, Crawford JD, Szebeni K, Chandley MJ, Burgess KC, Miller C, Bakkalbasi E, Brown RW. Antidepressant-Like Actions of Inhibitors of Poly(ADP-Ribose) Polymerase in Rodent Models. Int J Neuropsychopharmacol 2017; 20:994-1004. [PMID: 29016792 PMCID: PMC5716178 DOI: 10.1093/ijnp/pyx068] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/28/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Many patients suffering from depressive disorders are refractory to treatment with currently available antidepressant medications, while many more exhibit only a partial response. These factors drive research to discover new pharmacological approaches to treat depression. Numerous studies demonstrate evidence of inflammation and elevated oxidative stress in major depression. Recently, major depression has been shown to be associated with elevated levels of DNA oxidation in brain cells, accompanied by increased gene expression of the nuclear base excision repair enzyme, poly(ADP-ribose) polymerase-1. Given these findings and evidence that drugs that inhibit poly(ADP-ribose) polymerase-1 activity have antiinflammatory and neuroprotective properties, the present study was undertaken to examine the potential antidepressant properties of poly(ADP-ribose) polymerase inhibitors. METHODS Two rodent models, the Porsolt swim test and repeated exposure to psychological stressors, were used to test the poly(ADP-ribose) polymerase inhibitor, 3-aminobenzamide, for potential antidepressant activity. Another poly(ADP-ribose) polymerase inhibitor, 5-aminoisoquinolinone, was also tested. RESULTS Poly(ADP-ribose) polymerase inhibitors produced antidepressant-like effects in the Porsolt swim test, decreasing immobility time, and increasing latency to immobility, similar to the effects of fluoxetine. In addition, 3-aminobenzamide treatment increased sucrose preference and social interaction times relative to vehicle-treated control rats following repeated exposure to combined social defeat and unpredictable stress, mediating effects similar to fluoxetine treatment. CONCLUSIONS The poly(ADP-ribose) polymerase inhibitors 3-aminobenzamide and 5-aminoisoquinolinone exhibit antidepressant-like activity in 2 rodent stress models and uncover poly(ADP-ribose) polymerase as a unique molecular target for the potential development of a novel class of antidepressants.
Collapse
Affiliation(s)
- Gregory A Ordway
- Department of Biomedical Sciences (Drs Ordway and Szebeni, Ms Hernandez, Drs Crawford and Szebeni, Ms Burgess, and Dr Brown) and Department of Psychiatry and Behavioral Sciences (Dr Ordway), James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee; Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson City, Tennessee (Dr Chandley); DS Therapeutics, Houston, Texas (Dr Miller and Dr Bakkalbasi),Correspondence: Gregory A. Ordway, PhD, East Tennessee State University, PO Box 70577, Johnson City, 37614 ()
| | - Attila Szebeni
- Department of Biomedical Sciences (Drs Ordway and Szebeni, Ms Hernandez, Drs Crawford and Szebeni, Ms Burgess, and Dr Brown) and Department of Psychiatry and Behavioral Sciences (Dr Ordway), James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee; Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson City, Tennessee (Dr Chandley); DS Therapeutics, Houston, Texas (Dr Miller and Dr Bakkalbasi)
| | - Liza J Hernandez
- Department of Biomedical Sciences (Drs Ordway and Szebeni, Ms Hernandez, Drs Crawford and Szebeni, Ms Burgess, and Dr Brown) and Department of Psychiatry and Behavioral Sciences (Dr Ordway), James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee; Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson City, Tennessee (Dr Chandley); DS Therapeutics, Houston, Texas (Dr Miller and Dr Bakkalbasi)
| | - Jessica D Crawford
- Department of Biomedical Sciences (Drs Ordway and Szebeni, Ms Hernandez, Drs Crawford and Szebeni, Ms Burgess, and Dr Brown) and Department of Psychiatry and Behavioral Sciences (Dr Ordway), James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee; Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson City, Tennessee (Dr Chandley); DS Therapeutics, Houston, Texas (Dr Miller and Dr Bakkalbasi)
| | - Katalin Szebeni
- Department of Biomedical Sciences (Drs Ordway and Szebeni, Ms Hernandez, Drs Crawford and Szebeni, Ms Burgess, and Dr Brown) and Department of Psychiatry and Behavioral Sciences (Dr Ordway), James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee; Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson City, Tennessee (Dr Chandley); DS Therapeutics, Houston, Texas (Dr Miller and Dr Bakkalbasi)
| | - Michelle J Chandley
- Department of Biomedical Sciences (Drs Ordway and Szebeni, Ms Hernandez, Drs Crawford and Szebeni, Ms Burgess, and Dr Brown) and Department of Psychiatry and Behavioral Sciences (Dr Ordway), James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee; Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson City, Tennessee (Dr Chandley); DS Therapeutics, Houston, Texas (Dr Miller and Dr Bakkalbasi)
| | - Katherine C Burgess
- Department of Biomedical Sciences (Drs Ordway and Szebeni, Ms Hernandez, Drs Crawford and Szebeni, Ms Burgess, and Dr Brown) and Department of Psychiatry and Behavioral Sciences (Dr Ordway), James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee; Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson City, Tennessee (Dr Chandley); DS Therapeutics, Houston, Texas (Dr Miller and Dr Bakkalbasi)
| | - Corwin Miller
- Department of Biomedical Sciences (Drs Ordway and Szebeni, Ms Hernandez, Drs Crawford and Szebeni, Ms Burgess, and Dr Brown) and Department of Psychiatry and Behavioral Sciences (Dr Ordway), James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee; Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson City, Tennessee (Dr Chandley); DS Therapeutics, Houston, Texas (Dr Miller and Dr Bakkalbasi)
| | - Erol Bakkalbasi
- Department of Biomedical Sciences (Drs Ordway and Szebeni, Ms Hernandez, Drs Crawford and Szebeni, Ms Burgess, and Dr Brown) and Department of Psychiatry and Behavioral Sciences (Dr Ordway), James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee; Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson City, Tennessee (Dr Chandley); DS Therapeutics, Houston, Texas (Dr Miller and Dr Bakkalbasi)
| | - Russell W Brown
- Department of Biomedical Sciences (Drs Ordway and Szebeni, Ms Hernandez, Drs Crawford and Szebeni, Ms Burgess, and Dr Brown) and Department of Psychiatry and Behavioral Sciences (Dr Ordway), James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee; Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson City, Tennessee (Dr Chandley); DS Therapeutics, Houston, Texas (Dr Miller and Dr Bakkalbasi)
| |
Collapse
|