1
|
Gary AA, Prislovsky A, Tovar A, Locatelli E, Felix ER, Stephenson D, Chalfant CE, Lai J, Kim C, Mandal N, Galor A. Lipids from ocular meibum and tears may serve as biomarkers for depression and post-traumatic stress disorder. Clin Exp Ophthalmol 2024; 52:516-527. [PMID: 38146655 PMCID: PMC11199378 DOI: 10.1111/ceo.14343] [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: 08/18/2023] [Revised: 11/14/2023] [Accepted: 12/01/2023] [Indexed: 12/27/2023]
Abstract
BACKGROUND There is a need to develop biomarkers for diagnosis and prediction of treatment responses in depression and post-traumatic stress disorder (PTSD). METHODS Cross-sectional study examining correlations between tear inflammatory proteins, meibum and tear sphingolipids, and symptoms of depression and PTSD-associated anxiety. Ninety individuals filled depression (Patient Health Questionnaire 9, PHQ-9) and PTSD-associated anxiety (PTSD Checklist-Military Version, PCL-M) questionnaires. In 40 patients, a multiplex assay system was used to quantify 23 inflammatory proteins in tears. In a separate group of 50 individuals, liquid chromatography-mass spectrometry was performed on meibum and tears to quantify 34 species of sphingolipids, encompassing ceramides, monohexosyl ceramides and sphingomyelins. RESULTS The mean age of the population was 59.4 ± 11.0 years; 89.0% self-identified as male, 34.4% as White, 64.4% as Black, and 16.7% as Hispanic. The mean PHQ-9 score was 11.1 ± 7.6, and the mean PCL-M score was 44.3 ± 19.1. Symptoms of depression and PTSD-associated anxiety were highly correlated (ρ =0.75, p < 0.001). Both PHQ9 and PCL-M scores negatively correlated with multiple sphingolipid species in meibum and tears. In multivariable models, meibum Monohexosyl Ceramide 26:0 (pmol), tear Ceramide 16:0 (mol%), meibum Monohexosyl Ceramide 16:0 (mol%), and tear Ceramide 26:1 (mol%) remained associated with depression and meibum Monohexosyl Ceramide 16:0 (mol%), meibum Monohexosyl Ceramide 26:0 (pmol), tear Sphingomyelin 20:0 (mol%), and tear Sphingosine-1-Phosphate (mol%) remained associated with PTSD-associated anxiety. CONCLUSIONS Certain meibum and tear sphingolipid species were related to mental health indices. These interactions present opportunities for innovative diagnostic and therapeutic approaches for mental health disorders.
Collapse
Affiliation(s)
- Ashlyn A. Gary
- Miller School of Medicine, University of Miami, Miami, FL, USA
| | | | - Arianna Tovar
- Surgical Services, Miami Veterans Affairs Medical Center, Miami, FL, USA
| | - Elyana Locatelli
- Surgical Services, Miami Veterans Affairs Medical Center, Miami, FL, USA
- Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA
| | - Elizabeth R. Felix
- Research Service, Miami Veterans Affairs Medical Center, Miami, FL, USA
- Department of Physical Medicine & Rehabilitation, University of Miami, Miami, FL, USA
| | - Daniel Stephenson
- Departments of Medicine and Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Charles E. Chalfant
- Departments of Medicine and Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - James Lai
- Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Colin Kim
- Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Nawajes Mandal
- Memphis VA Medical Center, Memphis, TN, USA
- Depts. of Ophthalmology, Anatomy and Neurobiology and Pharmaceutical Sciences, University of Tennessee Health Sciences Center, Hamilton Eye Institute, Memphis, TN, USA
| | - Anat Galor
- Surgical Services, Miami Veterans Affairs Medical Center, Miami, FL, USA
- Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA
| |
Collapse
|
2
|
Tyrtyshnaia A, Manzhulo O, Manzhulo I. Synaptamide Ameliorates Hippocampal Neurodegeneration and Glial Activation in Mice with Traumatic Brain Injury. Int J Mol Sci 2023; 24:10014. [PMID: 37373162 DOI: 10.3390/ijms241210014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Traumatic brain injury (TBI) is a major concern for public health worldwide, affecting 55 million people and being the leading cause of death and disability. To improve the outcomes and effectiveness of treatment for these patients, we conducted a study on the potential therapeutic use of N-docosahexaenoylethanolamine (synaptamide) in mice using the weight-drop injury (WDI) TBI model. Our study focused on exploring synaptamide's effects on neurodegeneration processes and changes in neuronal and glial plasticity. Our findings showed that synaptamide could prevent TBI-associated working memory decline and neurodegenerative changes in the hippocampus, and it could alleviate decreased adult hippocampal neurogenesis. Furthermore, synaptamide regulated the production of astro- and microglial markers during TBI, promoting the anti-inflammatory transformation of the microglial phenotype. Additional effects of synaptamide in TBI include stimulating antioxidant and antiapoptotic defense, leading to the downregulation of the Bad pro-apoptotic marker. Our data suggest that synaptamide has promising potential as a therapeutic agent to prevent the long-term neurodegenerative consequences of TBI and improve the quality of life.
Collapse
Affiliation(s)
- Anna Tyrtyshnaia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Palchevskogo Str. 17, Vladivostok 690041, Russia
| | - Olga Manzhulo
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Palchevskogo Str. 17, Vladivostok 690041, Russia
| | - Igor Manzhulo
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Palchevskogo Str. 17, Vladivostok 690041, Russia
| |
Collapse
|
3
|
Hosseini E, Kianifard D. Effect of prenatal stress and extremely low-frequency electromagnetic fields on anxiety-like behavior in female rats: With an emphasis on prefrontal cortex and hippocampus. Brain Behav 2023; 13:e2949. [PMID: 36942730 PMCID: PMC10097060 DOI: 10.1002/brb3.2949] [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: 05/04/2022] [Revised: 09/03/2022] [Accepted: 01/23/2023] [Indexed: 03/23/2023] Open
Abstract
OBJECTIVE Prenatal stress (PS) is a problematic situation resulting in psychological implications such as social anxiety. Ubiquitous extremely low-frequency electromagnetic fields (ELF-EMF) have been confirmed as a potential physiological stressor; however, useful neuroregenerative effect of these types of electromagnetic fields has also frequently been reported. The aim of the present study was to survey the interaction of PS and ELF-EMF on anxiety-like behavior. METHOD A total of 24 female rats 40 days of age were distributed into four groups of 6 rats each: control, stress (their mothers were exposed to stress), EMF (their mothers underwent to ELF-EMF), and EMF/stress (their mothers concurrently underwent to stress and ELF-EMF). The rats were assayed using elevated plus-maze and open field tests. RESULTS Expressions of the hippocampus GAP-43, BDNF, and caspase-3 (cas-3) were detected by immunohistochemistry in Cornu Ammonis 1 (CA1) and dentate gyrus (DG) of the hippocampus and prefrontal cortex (PFC). Anxiety-like behavior increased in all treatment groups. Rats in the EMF/stress group presented more serious anxiety-like behavior. In all treatment groups, upregulated expression of cas-3 was seen in PFC, DG, and CA1 and downregulated expression of BDNF and GAP-43 was seen in PFC and DG and the CA1. Histomorphological study showed vast neurodegenerative changes in the hippocampus and PFC. CONCLUSION The results showed ,female rats that underwent PS or/and EMF exhibited critical anxiety-like behavior and this process may be attributed to neurodegeneration in PFC and DG of the hippocampus and possibly decreased synaptic plasticity so-called areas.
Collapse
Affiliation(s)
- Ehsan Hosseini
- Faculty of Veterinary Medicine, Division of Physiology, Department of basic science, Urmia University, Urmia, Iran
| | - Davoud Kianifard
- Faculty of Veterinary Medicine, Department of Basic Sciences, University of Tabriz, Tabriz, Iran
| |
Collapse
|
4
|
Morén C, Treder N, Martínez-Pinteño A, Rodríguez N, Arbelo N, Madero S, Gómez M, Mas S, Gassó P, Parellada E. Systematic Review of the Therapeutic Role of Apoptotic Inhibitors in Neurodegeneration and Their Potential Use in Schizophrenia. Antioxidants (Basel) 2022; 11:2275. [PMID: 36421461 PMCID: PMC9686909 DOI: 10.3390/antiox11112275] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 09/15/2023] Open
Abstract
Schizophrenia (SZ) is a deleterious brain disorder affecting cognition, emotion and reality perception. The most widely accepted neurochemical-hypothesis is the imbalance of neurotransmitter-systems. Depleted GABAergic-inhibitory function might produce a regionally-located dopaminergic and glutamatergic-storm in the brain. The dopaminergic-release may underlie the positive psychotic-symptoms while the glutamatergic-release could prompt the primary negative symptoms/cognitive deficits. This may occur due to excessive synaptic-pruning during the neurodevelopmental stages of adolescence/early adulthood. Thus, although SZ is not a neurodegenerative disease, it has been suggested that exaggerated dendritic-apoptosis could explain the limited neuroprogression around its onset. This apoptotic nature of SZ highlights the potential therapeutic action of anti-apoptotic drugs, especially at prodromal stages. If dysregulation of apoptotic mechanisms underlies the molecular basis of SZ, then anti-apoptotic molecules could be a prodromal therapeutic option to halt or prevent SZ. In fact, risk alleles related in apoptotic genes have been recently associated to SZ and shared molecular apoptotic changes are common in the main neurodegenerative disorders and SZ. PRISMA-guidelines were considered. Anti-apoptotic drugs are commonly applied in classic neurodegenerative disorders with promising results. Despite both the apoptotic-hallmarks of SZ and the widespread use of anti-apoptotic targets in neurodegeneration, there is a strikingly scarce number of studies investigating anti-apoptotic approaches in SZ. We analyzed the anti-apoptotic approaches conducted in neurodegeneration and the potential applications of such anti-apoptotic therapies as a promising novel therapeutic strategy, especially during early stages.
Collapse
Affiliation(s)
- Constanza Morén
- Barcelona Clínic Schizophrenia Unit (BCSU), Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
- Clinical and Experimental Neuroscience Area, The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- U722 Group, Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Carlos III Health Institute, 28029 Madrid, Spain
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
| | - Nina Treder
- Faculty of Psychology and Neuroscience, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Albert Martínez-Pinteño
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
| | - Natàlia Rodríguez
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
| | - Néstor Arbelo
- Barcelona Clínic Schizophrenia Unit (BCSU), Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
| | - Santiago Madero
- Barcelona Clínic Schizophrenia Unit (BCSU), Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
| | - Marta Gómez
- Barcelona Clínic Schizophrenia Unit (BCSU), Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
- Department of Psychiatry, Servizo Galego de Saúde (SERGAS), 36001 Pontevedra, Spain
| | - Sergi Mas
- Clinical and Experimental Neuroscience Area, The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
| | - Patricia Gassó
- Clinical and Experimental Neuroscience Area, The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
| | - Eduard Parellada
- Barcelona Clínic Schizophrenia Unit (BCSU), Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
- Clinical and Experimental Neuroscience Area, The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
| |
Collapse
|
5
|
Osacka J, Kiss A, Pirnik Z. Possible involvement of apoptosis in the antipsychotics side effects: A minireview. Clin Exp Pharmacol Physiol 2022; 49:836-847. [PMID: 35575958 DOI: 10.1111/1440-1681.13671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 01/04/2022] [Accepted: 05/11/2022] [Indexed: 11/28/2022]
Abstract
Antipsychotics are used in the treatment of schizophrenia and other psychiatric disorders. Generally they are divided into typical and atypical ones, according to the fact that atypical antipychotics induce less side effects and are more effective in terms of social and cognitive improvements. Their pharmacological effects are mediated via broad range of receptors that consequently influence different cellular signaling pathways. Antipsychotics produce udesirable side effects that range from relatively minor to life-threatening ones. In vitro and in vivo studies have pointed to neurotoxic effect exerted by some antipsychotics and have shown that apoptosis might play role in some side effects induced by antipsyschotics, including tardive dyskinesia, weight gain, agranulocytosis, osteoporosis, myocarditis, etc. Although cumulative data have suggested safety of atypical antipsychotics use during pregnancy some of them have been shown to induce apoptotic neurodegenerative and structural changes in fetal brains with long-lasting impact on cognitive impairment of offsprings. Typical antipsychotics seem to be more cytotoxic than atypical ones. Recently, epidemiological studies have shown lower incidence of cancer in schizophrenic patients what suggest ability of antipsychotics to suppress risk of cancer development. Some antipsychotics have been reported to inhibit cancer cell proliferation and induce their apoptosis. Thus, antipsychotics apoptotic effect may be used as a tool in the treatmnet of some types of cancer, especially in combinatorial therapies. In this minireview, we focused on pro- and anti-apototic or "Dr. Jekyll and Mr. Hyde" effects of antipsychotics, which can be involved in their side effects, as well as their promising therapeutical indications. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Jana Osacka
- Biomedical Research Center, Slovak Academy of Sciences, Institute of Experimental Endocrinology, Dubravska cesta 9, Bratislava, Slovakia
| | - Alexander Kiss
- Biomedical Research Center, Slovak Academy of Sciences, Institute of Experimental Endocrinology, Dubravska cesta 9, Bratislava, Slovakia
| | - Zdenko Pirnik
- Biomedical Research Center, Slovak Academy of Sciences, Institute of Experimental Endocrinology, Dubravska cesta 9, Bratislava, Slovakia.,Institute of Physiology, Faculty of Medicine Comenius University in Bratislava, Sasinkova 2, Bratislava, Slovakia.,Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nam., 2Prague, Czech Republic
| |
Collapse
|
6
|
Roy J, Wong KY, Aquili L, Uddin MS, Heng BC, Tipoe GL, Wong KH, Fung ML, Lim LW. Role of melatonin in Alzheimer's disease: From preclinical studies to novel melatonin-based therapies. Front Neuroendocrinol 2022; 65:100986. [PMID: 35167824 DOI: 10.1016/j.yfrne.2022.100986] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 01/21/2022] [Accepted: 02/07/2022] [Indexed: 12/11/2022]
Abstract
Melatonin and novel melatonin-based therapies such as melatonin-containing hybrid molecules, melatonin analogues, and melatonin derivatives have been investigated as potential therapeutics against Alzheimer's disease (AD) pathogenesis. In this review, we examine the developmental trends of melatonin therapies for AD from 1997 to 2021. We then highlight the neuroprotective mechanisms of melatonin therapy derived from preclinical studies. These mechanisms include the alleviation of amyloid-related burden, neurofibrillary tangle accumulation, oxidative stress, neuroinflammation, apoptosis, mitochondrial dysfunction, and impaired neuroplasticity and neurotransmission. We further illustrate the beneficial effects of melatonin on behavior in animal models of AD. Next, we discuss the clinical effects of melatonin on sleep, cognition, behavior, psychiatric symptoms, electroencephalography findings, and molecular biomarkers in patients with mild cognitive impairment and AD. We then explore the effectiveness of novel melatonin-based therapies. Lastly, we discuss the limitations of current melatonin therapies for AD and suggest two emerging research themes for future study.
Collapse
Affiliation(s)
- Jaydeep Roy
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kan Yin Wong
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Luca Aquili
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; College of Science, Health, Engineering and Education, Discipline of Psychology, Murdoch University, Perth, Australia
| | - Md Sahab Uddin
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Boon Chin Heng
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; Peking University School of Stomatology, Beijing, China
| | - George Lim Tipoe
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kah Hui Wong
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; Department of Anatomy, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Man Lung Fung
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Lee Wei Lim
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
7
|
Wu X, Zhu L, Zhao Z, Xu B, Yang J, Long J, Su L. Application of machine learning in diagnostic value of mRNAs for bipolar disorder. Nord J Psychiatry 2022; 76:81-88. [PMID: 34156910 DOI: 10.1080/08039488.2021.1937311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
PURPOSE Bipolar disorder (BD) is a type of severe mental illness with symptoms of mania or depression, it is necessary to find out effective diagnostic biomarkers for BD due to diagnosing BD is based on clinical interviews without objective indicators. MATERIALS AND METHODS The mRNA expression levels of genes included PIK3R1, FYN, TP53, PRKCZ, PRKCB, and YWHAB in the peripheral blood of 43 patients with bipolar disorder and 47 healthy controls were detected. Machine learning methods included Artificial Neural Networks, Extreme Gradient Boosting, Random Forest, and Support Vector Machine were adopted to fit different gene combinations to evaluate diagnostic value for bipolar disorder. RESULTS The combination 'PIK3R1 + FYN' in the SVM model showed the best diagnostic value, with AUC, sensitivity, and specificity values of 0.951, 0.928, and 0.937, respectively. CONCLUSIONS The diagnostic efficiency for bipolar disorder was significantly improved by fitting PIK3R1 and FYN through the Support Vector Machine model.
Collapse
Affiliation(s)
- Xulong Wu
- School of Public Health, Guangxi Medical University, Guangxi, China
| | - Lulu Zhu
- School of Public Health, Guangxi Medical University, Guangxi, China
| | - Zhi Zhao
- School of Public Health, Guangxi Medical University, Guangxi, China
| | - Bingyi Xu
- School of Public Health, Guangxi Medical University, Guangxi, China
| | - Jialei Yang
- School of Public Health, Guangxi Medical University, Guangxi, China
| | - Jianxiong Long
- School of Public Health, Guangxi Medical University, Guangxi, China
| | - Li Su
- School of Public Health, Guangxi Medical University, Guangxi, China
| |
Collapse
|
8
|
Koga M, Nakagawa S, Sato A, Oka M, Makikhara K, Sakai Y, Toyomaki A, Sato M, Matsui M, Toda H, Kusumi I. Plasma fatty acid-binding protein 7 concentration correlates with depression/anxiety, cognition, and positive symptom in patients with schizophrenia. J Psychiatr Res 2021; 144:304-311. [PMID: 34715597 DOI: 10.1016/j.jpsychires.2021.10.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/08/2021] [Accepted: 10/19/2021] [Indexed: 10/20/2022]
Abstract
Because of the involvement of the brain in the pathophysiology of psychiatric disorders, obtaining information on the biochemical features that directly contribute to symptoms is challenging. The present study aimed to assess fatty acid-binding protein 7 (FABP7) expressed specifically in the brain and detectable in the peripheral blood and to investigate the correlation between blood FABP7 concentration and symptoms. We recruited 30, 29, and 35 patients with schizophrenia, bipolar disorder, and depression and evaluated using the Positive and Negative Syndrome Scale (PANSS), Young Mania Rating Scale (YMRS), and Hamilton Depression Rating Scale (HAMD-21), respectively. Plasma FABP7 concentrations correlated with PANSS scores (R2 = 0.3305, p < 0.001) but not with other scales. In the analysis of the relationship between five dimensions of schizophrenia symptoms derived from the PANSS 5-factor model and measured plasma FABP7 concentrations, severities of depression/anxiety, cognition, and positive symptom were significantly correlated with plasma FABP7 concentrations. Further molecular investigation of the functional and kinetic analyses of FABP7 is necessary to understand the relationship of this protein with schizophrenia pathology. Nevertheless, the present study suggests that FABP7 can be a biological indicator reflecting the pathogenesis of schizophrenia and has potential applications as a biomarker for diagnosis and symptom assessment.
Collapse
Affiliation(s)
- Minori Koga
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan; Department of Psychiatry, National Defense Medical College, Tokorozawa, Saitama, Japan.
| | - Shin Nakagawa
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan; Yamaguchi University Graduate School of Medicine Division of Neuropsychiatry, Department of Neuroscience, Japan
| | - Asumi Sato
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Matsuhiko Oka
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Keisuke Makikhara
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Yuri Sakai
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Atsuhito Toyomaki
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Mayumi Sato
- Department of Psychiatry, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Marie Matsui
- Department of Psychiatry, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hiroyuki Toda
- Department of Psychiatry, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Ichiro Kusumi
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| |
Collapse
|
9
|
Sharma VK, Singh TG, Singh S, Garg N, Dhiman S. Apoptotic Pathways and Alzheimer's Disease: Probing Therapeutic Potential. Neurochem Res 2021; 46:3103-3122. [PMID: 34386919 DOI: 10.1007/s11064-021-03418-7] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 12/17/2022]
Abstract
Apoptosis is an intrinsic biochemical, cellular process that regulates cell death and is crucial for cell survival, cellular homeostasis, and maintaining the optimum functional status. Apoptosis in a predetermined and programmed manner regulates several molecular events, including cell turnover, embryonic development, and immune system functions but may be the exclusive contributor to several disorders, including neurodegenerative manifestations, when it functions in an aberrant and disorganized manner. Alzheimer's disease (AD) is a fatal, chronic neurodegenerative disorder where apoptosis has a compelling and divergent role. The well-characterized pathological features of AD, including extracellular plaques of amyloid-beta, intracellular hyperphosphorylated tangles of tau protein (NFTs), inflammation, mitochondrial dysfunction, oxidative stress, and excitotoxic cell death, also instigate an abnormal apoptotic cascade in susceptible brain regions (cerebral cortex, hippocampus). The apoptotic players in these regions affect cellular organelles (mitochondria and endoplasmic reticulum), interact with trophic factors, and several pathways, including PI3K/AKT, JNK, MAPK, mTOR signalling. This dysregulated apoptotic cascade end with an abnormal neuronal loss which is a primary event that may precede the other events of AD progression and correlates well with the degree of dementia. The present review provides insight into the diverse and versatile apoptotic mechanisms that are indispensable for neuronal survival and constitute an integral part of the pathological progression of AD. Identification of potential targets (restoring apoptotic and antiapoptotic balance, caspases, TRADD, RIPK1, FADD, TNFα, etc.) may be valuable and advantageous to decide the fate of neurons and to develop potential therapeutics for treatment of AD.
Collapse
Affiliation(s)
- Vivek Kumar Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India.,Government College of Pharmacy, Rohru, District Shimla, Himachal Pradesh, 171207, India
| | | | - Shareen Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | - Nikhil Garg
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | - Sonia Dhiman
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| |
Collapse
|
10
|
Wanderoy S, Hees JT, Klesse R, Edlich F, Harbauer AB. Kill one or kill the many: interplay between mitophagy and apoptosis. Biol Chem 2020; 402:73-88. [PMID: 33544491 DOI: 10.1515/hsz-2020-0231] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/22/2020] [Indexed: 12/15/2022]
Abstract
Mitochondria are key players of cellular metabolism, Ca2+ homeostasis, and apoptosis. The functionality of mitochondria is tightly regulated, and dysfunctional mitochondria are removed via mitophagy, a specialized form of autophagy that is compromised in hereditary forms of Parkinson's disease. Through mitophagy, cells are able to cope with mitochondrial stress until the damage becomes too great, which leads to the activation of pro-apoptotic BCL-2 family proteins located on the outer mitochondrial membrane. Active pro-apoptotic BCL-2 proteins facilitate the release of cytochrome c from the mitochondrial intermembrane space (IMS) into the cytosol, committing the cell to apoptosis by activating a cascade of cysteinyl-aspartate specific proteases (caspases). We are only beginning to understand how the choice between mitophagy and the activation of caspases is determined on the mitochondrial surface. Intriguingly in neurons, caspase activation also plays a non-apoptotic role in synaptic plasticity. Here we review the current knowledge on the interplay between mitophagy and caspase activation with a special focus on the central nervous system.
Collapse
Affiliation(s)
- Simone Wanderoy
- Max Planck Institute for Neurobiology, Am Klopferspitz 18, D-82152Martinsried, Germany
| | - J Tabitha Hees
- Max Planck Institute for Neurobiology, Am Klopferspitz 18, D-82152Martinsried, Germany
| | - Ramona Klesse
- Institute for Biochemistry and Molecular Biology, University of Freiburg, D-79104Freiburg, Germany.,Faculty of Biology, University of Freiburg, D-79104Freiburg, Germany
| | - Frank Edlich
- Institute for Biochemistry and Molecular Biology, University of Freiburg, D-79104Freiburg, Germany
| | - Angelika B Harbauer
- Max Planck Institute for Neurobiology, Am Klopferspitz 18, D-82152Martinsried, Germany.,Technical University of Munich, Institute of Neuronal Cell Biology, Munich, Germany.,Munich Cluster for Systems Neurology, Munich, Germany
| |
Collapse
|
11
|
Qin X, Li L, Nie X, Niu Q. Effects of Chronic Aluminum Lactate Exposure on Neuronal Apoptosis and Hippocampal Synaptic Plasticity in Rats. Biol Trace Elem Res 2020; 197:571-579. [PMID: 31845204 DOI: 10.1007/s12011-019-02007-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 12/05/2019] [Indexed: 12/22/2022]
Abstract
This study investigated the effects of chronic aluminum exposure on apoptosis of hippocampal neurons, and synaptic plasticity in the hippocampus in rats. Rats were divided into the control, low-dose (L-Al), mid-dose (M-Al), and high-dose (H-Al) groups. After chronic exposure of aluminum, the Morris water maze (MWM) and open-field (OF) tests were performed to assess the behavioral performance. Electrophysiological measurements were conducted. Flow cytometry was used to assess the apoptotic processes. Quantitative real-time PCR and ELISA were performed to measure mRNA and protein expression levels of caspases. After 90 days of aluminum exposure, the aluminum contents in the brain of the rats were increased, with the increasing exposure dose. The MWM and OF tests showed that chronic exposure of aluminum significantly impaired the neurobehavior of rats. Moreover, after high-frequency stimulation (HFS), the average amplitudes of field excitatory postsynaptic potentials (fEPSPs) for the M-Al and H-Al groups were lower than the control group at 10, 20, 30, 40, 50, and 60 min. Furthermore, the apoptotic rates in the M-Al and H-Al groups were significantly higher than the control group. The qRT-PCR and ELISA showed that, compared with the control group, the mRNA and protein expression levels of caspases-3, -8, and -9 were significantly increased in the aluminum-treated groups compared with the control group. Long-term exposure to aluminum could induce the apoptosis of hippocampal neurons, damage the synaptic plasticity, and impair the learning and memory functions in rats. There might be a close relationship between the neuronal apoptosis and synaptic plasticity damage.
Collapse
Affiliation(s)
- Xiujun Qin
- Shanxi Provincial Key Laboratory of Environment and Health Impairment, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
- China Institute for Radiation Protection, Taiyuan, 030006, Shanxi, China
| | - Liang Li
- Shanxi Provincial Key Laboratory of Environment and Health Impairment, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Xiaohan Nie
- Shanxi Provincial Key Laboratory of Environment and Health Impairment, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Qiao Niu
- Shanxi Provincial Key Laboratory of Environment and Health Impairment, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
- Key Laboratory of Cellular Physiology of Education Ministry, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
12
|
Johnstone A, Mobley W. Local TrkB signaling: themes in development and neural plasticity. Cell Tissue Res 2020; 382:101-111. [DOI: 10.1007/s00441-020-03278-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/10/2020] [Indexed: 02/08/2023]
|
13
|
Genome-wide association study and polygenic risk score analysis of esketamine treatment response. Sci Rep 2020; 10:12649. [PMID: 32724131 PMCID: PMC7387452 DOI: 10.1038/s41598-020-69291-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/01/2020] [Indexed: 12/13/2022] Open
Abstract
To elucidate the genetic underpinnings of the antidepressant efficacy of S-ketamine (esketamine) nasal spray in major depressive disorder (MDD), we performed a genome-wide association study (GWAS) in cohorts of European ancestry (n = 527). This analysis was followed by a polygenic risk score approach to test for associations between genetic loading for psychiatric conditions, symptom profiles and esketamine efficacy. We identified a genome-wide significant locus in IRAK3 (p = 3.57 × 10–8, rs11465988, β = − 51.6, SE = 9.2) and a genome-wide significant gene-level association in NME7 (p = 1.73 × 10–6) for esketamine efficacy (i.e. percentage change in symptom severity score compared to baseline). Additionally, the strongest association with esketamine efficacy identified in the polygenic score analysis was from the genetic loading for depressive symptoms (p = 0.001, standardized coefficient β = − 3.1, SE = 0.9), which did not reach study-wide significance. Pathways relevant to neuronal and synaptic function, immune signaling, and glucocorticoid receptor/stress response showed enrichment among the suggestive GWAS signals.
Collapse
|
14
|
Coccia E, Planells-Ferrer L, Badillos-Rodríguez R, Pascual M, Segura MF, Fernández-Hernández R, López-Soriano J, Garí E, Soriano E, Barneda-Zahonero B, Moubarak RS, Pérez-García MJ, Comella JX. SIVA-1 regulates apoptosis and synaptic function by modulating XIAP interaction with the death receptor antagonist FAIM-L. Cell Death Dis 2020; 11:82. [PMID: 32015347 PMCID: PMC6997380 DOI: 10.1038/s41419-020-2282-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 12/15/2022]
Abstract
The long isoform of Fas apoptosis inhibitory molecule (FAIM-L) is a neuron-specific death receptor antagonist that modulates apoptotic cell death and mechanisms of neuronal plasticity. FAIM-L exerts its antiapoptotic action by binding to X-linked inhibitor of apoptosis protein (XIAP), an inhibitor of caspases, which are the main effectors of apoptosis. XIAP levels are regulated by the ubiquitin-proteasome pathway. FAIM-L interaction with XIAP prevents the ubiquitination and degradation of the latter, thereby allowing it to inhibit caspase activation. This interaction also modulates non-apoptotic functions of caspases, such as the endocytosis of AMPA receptor (AMPAR) in hippocampal long-term depression (LTD). The molecular mechanism of action exerted by FAIM-L is unclear since the consensus binding motifs are still unknown. Here, we performed a two-hybrid screening to discover novel FAIM-L-interacting proteins. We found a functional interaction of SIVA-1 with FAIM-L. SIVA-1 is a proapoptotic protein that has the capacity to interact with XIAP. We describe how SIVA-1 regulates FAIM-L function by disrupting the interaction of FAIM-L with XIAP, thereby promoting XIAP ubiquitination, caspase-3 activation and neuronal death. Furthermore, we report that SIVA-1 plays a role in receptor internalization in synapses. SIVA-1 is upregulated upon chemical LTD induction, and it modulates AMPAR internalization via non-apoptotic activation of caspases. In summary, our findings uncover SIVA-1 as new functional partner of FAIM-L and demonstrate its role as a regulator of caspase activity in synaptic function.
Collapse
Affiliation(s)
- Elena Coccia
- Cell Signaling and Apoptosis Group, Vall d'Hebron Research Institute (VHIR), 08035, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain.,Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, 08031, Bellaterra, Spain
| | - Laura Planells-Ferrer
- Cell Signaling and Apoptosis Group, Vall d'Hebron Research Institute (VHIR), 08035, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain.,Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, 08031, Bellaterra, Spain
| | - Raquel Badillos-Rodríguez
- Cell Signaling and Apoptosis Group, Vall d'Hebron Research Institute (VHIR), 08035, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain.,Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, 08031, Bellaterra, Spain
| | - Marta Pascual
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain.,Institut de Neurociències, Universitat de Barcelona, Bellaterra, Spain.,Department of Cell Biology, Physiology and Immunology, Institut de Neurociències, Universitat de Barcelona, 08031, Barcelona, Spain
| | - Miguel F Segura
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute (VHIR)-UAB, 08035, Barcelona, Spain
| | - Rita Fernández-Hernández
- Cell Cycle Laboratory, Institut de Recerca Biomèdica de Lleida (IRBLleida), and Departament de Ciències Mèdiques Bàsiques; Facultat de Medicina, Universitat de Lleida, 25198, Lleida, Catalonia, Spain
| | - Joaquin López-Soriano
- Cell Signaling and Apoptosis Group, Vall d'Hebron Research Institute (VHIR), 08035, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain.,Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, 08031, Bellaterra, Spain
| | - Eloi Garí
- Cell Cycle Laboratory, Institut de Recerca Biomèdica de Lleida (IRBLleida), and Departament de Ciències Mèdiques Bàsiques; Facultat de Medicina, Universitat de Lleida, 25198, Lleida, Catalonia, Spain
| | - Eduardo Soriano
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain.,Institut de Neurociències, Universitat de Barcelona, Bellaterra, Spain.,Department of Cell Biology, Physiology and Immunology, Institut de Neurociències, Universitat de Barcelona, 08031, Barcelona, Spain.,ICREA Academia, Barcelona, Spain
| | - Bruna Barneda-Zahonero
- Cell Signaling and Apoptosis Group, Vall d'Hebron Research Institute (VHIR), 08035, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain.,Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, 08031, Bellaterra, Spain
| | - Rana S Moubarak
- Cell Signaling and Apoptosis Group, Vall d'Hebron Research Institute (VHIR), 08035, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain.,Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, 08031, Bellaterra, Spain.,Department of Pathology, NYU Langone Health, New York, 10016, NY, USA
| | - M Jose Pérez-García
- Cell Signaling and Apoptosis Group, Vall d'Hebron Research Institute (VHIR), 08035, Barcelona, Spain. .,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain. .,Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, 08031, Bellaterra, Spain.
| | - Joan X Comella
- Cell Signaling and Apoptosis Group, Vall d'Hebron Research Institute (VHIR), 08035, Barcelona, Spain. .,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain. .,Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, 08031, Bellaterra, Spain.
| |
Collapse
|
15
|
Chen X, Jiang Y, Wang J, Liu Y, Xiao M, Song C, Bai Y, Yinuo Han N, Han F. Synapse impairment associated with enhanced apoptosis in post-traumatic stress disorder. Synapse 2019; 74:e22134. [PMID: 31562782 DOI: 10.1002/syn.22134] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 09/17/2019] [Accepted: 09/20/2019] [Indexed: 12/31/2022]
Abstract
Synapse impairment is associated with post-traumatic stress disorder (PTSD), which is characterized by enhanced apoptosis in the hippocampus, amygdala, and other brain regions. However, there are no detailed studies on the relationship between apoptosis and synaptic connectivity in PTSD. In this review, we discuss results from various studies describing the synaptic changes observed in the PTSD brain. A decreased number of dendrites/spines or increased number of immature spines in the hippocampus, medial prefrontal cortex, and other brain regions has been reported. Studies on axon guidance, myelination, and the cytoskeleton suggest that PTSD may involve axon overgrowth and overbranching. Apoptosis affects synapse formation; low levels of caspase maintain the balance between growth cone attraction and repulsion and inhibit axon elongation. PTSD enhances neuronal apoptosis through caspase activation, which disrupts the balance between growth cone attraction and repulsion and alters growth cone trajectory, leading to axon mistargeting. Meanwhile, caspase activation induces dendritic pruning and dendrite degeneration. These events contribute to the formation of fewer and aberrant synapses, which is associated with enhanced apoptosis in PTSD.
Collapse
Affiliation(s)
- Xinzhao Chen
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Yifan Jiang
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Jiayu Wang
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Yishu Liu
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Menglei Xiao
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Congshan Song
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Yu Bai
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Nancy Yinuo Han
- Faculty of Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Fang Han
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| |
Collapse
|
16
|
Imbriani P, Tassone A, Meringolo M, Ponterio G, Madeo G, Pisani A, Bonsi P, Martella G. Loss of Non-Apoptotic Role of Caspase-3 in the PINK1 Mouse Model of Parkinson's Disease. Int J Mol Sci 2019; 20:ijms20143407. [PMID: 31336695 PMCID: PMC6678522 DOI: 10.3390/ijms20143407] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/04/2019] [Accepted: 07/09/2019] [Indexed: 11/23/2022] Open
Abstract
Caspases are a family of conserved cysteine proteases that play key roles in multiple cellular processes, including programmed cell death and inflammation. Recent evidence shows that caspases are also involved in crucial non-apoptotic functions, such as dendrite development, axon pruning, and synaptic plasticity mechanisms underlying learning and memory processes. The activated form of caspase-3, which is known to trigger widespread damage and degeneration, can also modulate synaptic function in the adult brain. Thus, in the present study, we tested the hypothesis that caspase-3 modulates synaptic plasticity at corticostriatal synapses in the phosphatase and tensin homolog (PTEN) induced kinase 1 (PINK1) mouse model of Parkinson’s disease (PD). Loss of PINK1 has been previously associated with an impairment of corticostriatal long-term depression (LTD), rescued by amphetamine-induced dopamine release. Here, we show that caspase-3 activity, measured after LTD induction, is significantly decreased in the PINK1 knockout model compared with wild-type mice. Accordingly, pretreatment of striatal slices with the caspase-3 activator α-(Trichloromethyl)-4-pyridineethanol (PETCM) rescues a physiological LTD in PINK1 knockout mice. Furthermore, the inhibition of caspase-3 prevents the amphetamine-induced rescue of LTD in the same model. Our data support a hormesis-based double role of caspase-3; when massively activated, it induces apoptosis, while at lower level of activation, it modulates physiological phenomena, like the expression of corticostriatal LTD. Exploring the non-apoptotic activation of caspase-3 may contribute to clarify the mechanisms involved in synaptic failure in PD, as well as in view of new potential pharmacological targets.
Collapse
Affiliation(s)
- Paola Imbriani
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Annalisa Tassone
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Maria Meringolo
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Giulia Ponterio
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Graziella Madeo
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Antonio Pisani
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy.
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
| | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
| |
Collapse
|
17
|
TP53 Polymorphism Contributes to the Susceptibility to Bipolar Disorder but Not to Schizophrenia in the Chinese Han Population. J Mol Neurosci 2019; 68:679-687. [DOI: 10.1007/s12031-019-01330-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/25/2019] [Indexed: 12/15/2022]
|
18
|
The Role of Apoptotic Signaling in Axon Guidance. J Dev Biol 2018; 6:jdb6040024. [PMID: 30340315 PMCID: PMC6316149 DOI: 10.3390/jdb6040024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 10/16/2018] [Accepted: 10/16/2018] [Indexed: 12/28/2022] Open
Abstract
Navigating growth cones are exposed to multiple signals simultaneously and have to integrate competing cues into a coherent navigational response. Integration of guidance cues is traditionally thought to occur at the level of cytoskeletal dynamics. Drosophila studies indicate that cells exhibit a low level of continuous caspase protease activation, and that axon guidance cues can activate or suppress caspase activity. We base a model for axon guidance on these observations. By analogy with other systems in which caspase signaling has non-apoptotic functions, we propose that caspase signaling can either reinforce repulsion or negate attraction in response to external guidance cues by cleaving cytoskeletal proteins. Over the course of an entire trajectory, incorrectly navigating axons may pass the threshold for apoptosis and be eliminated, whereas axons making correct decisions will survive. These observations would also explain why neurotrophic factors can act as axon guidance cues and why axon guidance systems such as Slit/Robo signaling may act as tumor suppressors in cancer.
Collapse
|
19
|
Hess JL, Akutagava-Martins GC, Patak JD, Glatt SJ, Faraone SV. Why is there selective subcortical vulnerability in ADHD? Clues from postmortem brain gene expression data. Mol Psychiatry 2018; 23:1787-1793. [PMID: 29180674 PMCID: PMC6985986 DOI: 10.1038/mp.2017.242] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/17/2017] [Accepted: 08/24/2017] [Indexed: 01/25/2023]
Abstract
Sub-cortical volumetric differences were associated with attention-deficit/hyperactivity disorder (ADHD) in a recent multi-site, mega-analysis of 1713 ADHD persons and 1529 controls. As there was a wide range of effect sizes among the sub-cortical volumes, it is possible that selective neuronal vulnerability has a role in these volumetric losses. To address this possibility, we used data from Allen Brain Atlas to investigate variability in gene expression profiles between subcortical regions of typically developing brains. We tested the hypothesis that the expression of genes in a set of curated ADHD candidate genes and five a priori selected, biological pathways would be associated with the Enhancing NeuroImaging Genetics through Meta-Analysis (ENIGMA) findings. Across the subcortical regions studied by ENIGMA, gene expression profiles for three pathways were significantly correlated with ADHD-associated volumetric reductions: apoptosis, oxidative stress and autophagy. These correlations were strong and significant for children with ADHD, but not for adults. Although preliminary, these data suggest that variability of structural brain anomalies in ADHD can be explained, in part, by the differential vulnerability of these regions to mechanisms mediating apoptosis, oxidative stress and autophagy.
Collapse
Affiliation(s)
- J L Hess
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY, USA
| | - G C Akutagava-Martins
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY, USA
| | - J D Patak
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - S J Glatt
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY, USA
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - S V Faraone
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY, USA.
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA.
- K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, University of Bergen, Bergen, Norway.
| |
Collapse
|
20
|
Samokhina E, Samokhin A. Neuropathological profile of the pentylenetetrazol (PTZ) kindling model. Int J Neurosci 2018; 128:1086-1096. [DOI: 10.1080/00207454.2018.1481064] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- E. Samokhina
- Russian Academy of Sciences, Institute of Theoretical and Experimental Biophysics, Pushchino, Russia
| | - Alexander Samokhin
- Russian Academy of Sciences, Institute of Cell Biophysics, Pushchino, Russia
| |
Collapse
|
21
|
Notch signaling and neuronal death in stroke. Prog Neurobiol 2018; 165-167:103-116. [PMID: 29574014 DOI: 10.1016/j.pneurobio.2018.03.002] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 02/08/2018] [Accepted: 03/20/2018] [Indexed: 12/18/2022]
Abstract
Ischemic stroke is a leading cause of morbidity and death, with the outcome largely determined by the amount of hypoxia-related neuronal death in the affected brain regions. Cerebral ischemia and hypoxia activate the Notch1 signaling pathway and four prominent interacting pathways (NF-κB, p53, HIF-1α and Pin1) that converge on a conserved DNA-associated nuclear multi-protein complex, which controls the expression of genes that can determine the fate of neurons. When neurons experience a moderate level of ischemic insult, the nuclear multi-protein complex up-regulates adaptive stress response genes encoding proteins that promote neuronal survival, but when ischemia is more severe the nuclear multi-protein complex induces genes encoding proteins that trigger and execute a neuronal death program. We propose that the nuclear multi-protein transcriptional complex is a molecular mediator of neuronal hormesis and a target for therapeutic intervention in stroke.
Collapse
|
22
|
Plant N. Can a systems approach produce a better understanding of mood disorders? Biochim Biophys Acta Gen Subj 2016; 1861:3335-3344. [PMID: 27565355 DOI: 10.1016/j.bbagen.2016.08.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 07/29/2016] [Accepted: 08/22/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND One in twenty-five people suffer from a mood disorder. Current treatments are sub-optimal with poor patient response and uncertain modes-of-action. There is thus a need to better understand underlying mechanisms that determine mood, and how these go wrong in affective disorders. Systems biology approaches have yielded important biological discoveries for other complex diseases such as cancer, and their potential in affective disorders will be reviewed. SCOPE OF REVIEW This review will provide a general background to affective disorders, plus an outline of experimental and computational systems biology. The current application of these approaches in understanding affective disorders will be considered, and future recommendations made. MAJOR CONCLUSIONS Experimental systems biology has been applied to the study of affective disorders, especially at the genome and transcriptomic levels. However, data generation has been slowed by a lack of human tissue or suitable animal models. At present, computational systems biology has only be applied to understanding affective disorders on a few occasions. These studies provide sufficient novel biological insight to motivate further use of computational biology in this field. GENERAL SIGNIFICANCE In common with many complex diseases much time and money has been spent on the generation of large-scale experimental datasets. The next step is to use the emerging computational approaches, predominantly developed in the field of oncology, to leverage the most biological insight from these datasets. This will lead to the critical breakthroughs required for more effective diagnosis, stratification and treatment of affective disorders.
Collapse
Affiliation(s)
- Nick Plant
- School of Bioscience and Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7XH, UK.
| |
Collapse
|
23
|
Sandrow-Feinberg HR, Houlé JD. Exercise after spinal cord injury as an agent for neuroprotection, regeneration and rehabilitation. Brain Res 2015; 1619:12-21. [PMID: 25866284 PMCID: PMC4540698 DOI: 10.1016/j.brainres.2015.03.052] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 03/27/2015] [Accepted: 03/31/2015] [Indexed: 12/17/2022]
Abstract
Spinal cord injury (SCI) is a traumatic event from which there is limited recovery of function, despite the best efforts of many investigators to devise realistic therapeutic treatments. Partly this is due to the multifaceted nature of SCI, where there is considerable disarray and dysfunction secondary to the initial injury. Contributing to this secondary degeneration is neurotoxicity, vascular dysfunction, glial scarring, neuroinflammation, apoptosis and demyelination. It seems logical that addressing the need for neuroprotection, regeneration and rehabilitation will require different treatment strategies that may be applied at varied stages of the post-injury response. Here we focus on a single strategy, exercise/physical training, which appears to have multiple applications and benefits for an acute or chronic SCI. Exercise has been demonstrated to be advantageous at cellular and biochemical levels, as well as being of benefit for the whole animal or human subject. Data from our lab and others will be discussed to further elucidate the many positive aspects of implementing exercise following injury and to suggest that rehabilitation is not the sole target of a training regimen following SCI. This article is part of a Special Issue entitled SI: Spinal cord injury.
Collapse
Affiliation(s)
- Harra R Sandrow-Feinberg
- Department of Neurobiology and Anatomy, Spinal Cord Research Center, Drexel University College of Medicine, 2900 Queen Lane, PA 19129, Philadelphia, United States
| | - John D Houlé
- Department of Neurobiology and Anatomy, Spinal Cord Research Center, Drexel University College of Medicine, 2900 Queen Lane, PA 19129, Philadelphia, United States.
| |
Collapse
|
24
|
Płóciennik A, Prendecki M, Zuba E, Siudzinski M, Dorszewska J. Activated Caspase-3 and Neurodegeneration and Synaptic Plasticity in Alzheimer’s Disease. ACTA ACUST UNITED AC 2015. [DOI: 10.4236/aad.2015.43007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
25
|
Rossi S, Motta C, Studer V, Macchiarulo G, Volpe E, Barbieri F, Ruocco G, Buttari F, Finardi A, Mancino R, Weiss S, Battistini L, Martino G, Furlan R, Drulovic J, Centonze D. Interleukin-1β causes excitotoxic neurodegeneration and multiple sclerosis disease progression by activating the apoptotic protein p53. Mol Neurodegener 2014; 9:56. [PMID: 25495224 PMCID: PMC4292815 DOI: 10.1186/1750-1326-9-56] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 12/03/2014] [Indexed: 12/16/2022] Open
Abstract
Background Understanding how inflammation causes neuronal damage is of paramount importance in multiple sclerosis (MS) and in other neurodegenerative diseases. Here we addressed the role of the apoptotic cascade in the synaptic abnormalities and neuronal loss caused by the proinflammatory cytokines interleukin-1β (IL-1β) and tumor necrosis factor (TNF-α) in brain tissues, and disease progression caused by inflammation in relapsing-remitting MS (RRMS) patients. Results The effect of IL-1β, but not of TNF-α, on glutamate-mediated excitatory postsynaptic currents was blocked by pifithrin-α (PFT), inhibitor of p53. The protein kinase C (PKC)/transient receptor potential vanilloid 1 (TRPV1) pathway was involved in IL-1β-p53 interaction at glutamatergic synapses, as pharmacological modulation of this inflammation-relevant molecular pathway affected PFT effects on the synaptic action of IL-1β. IL-1β-induced neuronal swelling was also blocked by PFT, and IL-1β increased the expression of p21, a canonical downstream target of activated p53. Consistent with these in vitro results, the Pro/Pro genotype of p53, associated with low efficiency of transcription of p53-regulated genes, abrogated the association between IL-1β cerebrospinal fluid (CSF) levels and disability progression in RRMS patients. The interaction between p53 and CSF IL-1β was also evaluated at the optical coherence tomography (OCT), showing that IL-1β-driven neurodegenerative damage, causing alterations of macular volume and of retinal nerve fibre layer thickness, was modulated by the p53 genotype. Conclusions Inflammatory synaptopathy and neurodegeneration caused by IL-1β in RRMS patients involve the apoptotic cascade. Targeting IL-1β-p53 interaction might result in significant neuroprotection in MS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Diego Centonze
- Clinica Neurologica, Dipartimento di Medicina dei Sistemi, Università Tor Vergata, Via Montpellier 1, 00133 Rome, Italy.
| |
Collapse
|
26
|
Fan W, Dai Y, Xu H, Zhu X, Cai P, Wang L, Sun C, Hu C, Zheng P, Zhao BQ. Caspase-3 modulates regenerative response after stroke. Stem Cells 2014; 32:473-86. [PMID: 23939807 DOI: 10.1002/stem.1503] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 07/28/2013] [Accepted: 07/31/2013] [Indexed: 12/27/2022]
Abstract
Stroke is a leading cause of long-lasting disability in humans. However, currently there are still no effective therapies available for promoting stroke recovery. Recent studies have shown that the adult brain has the capacity to regenerate neurons after stroke. Although this neurogenic response may be functionally important for brain repair after injury, the mechanisms underlying stroke-induced neurogenesis are not known. Caspase-3 is a major executioner and has been identified as a key mediator of neuronal death in the acute stage of stroke. Recently, however, accumulating data indicate that caspase-3 also participates in various biological processes that do not cause cell death. Here, we show that cleaved caspase-3 was increased in newborn neuronal precursor cells (NPCs) in the subventricular zone (SVZ) and the dentate gyrus during the period of stroke recovery, with no evidence of apoptosis. We observed that cleaved caspase-3 was expressed by NPCs and limited its self-renewal without triggering apoptosis in cultured NPCs from the SVZ of ischemic mice. Moreover, we revealed that caspase-3 negatively regulated the proliferation of NPCs through reducing the phosphorylation of Akt. Importantly, we demonstrated that peptide inhibition of caspase-3 activity significantly promoted the proliferation and migration of SVZ NPCs and resulted in a significant increase in subsequent neuronal regeneration and functional recovery after stroke. Together, our data identify a previously unknown caspase-3-dependent mechanism that constrains stroke-induced endogenous neurogenesis and should revitalize interest in targeting caspase-3 for treatment of stroke.
Collapse
Affiliation(s)
- Wenying Fan
- State Key Laboratory of Medical Neurobiology, Shanghai Medical College and Institutes of Brain Science, Fudan University, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Kudryashova IV, Onufriev MV, Gulyaeva NV. Caspase-3 and calpain: Differently directed involvement in presynaptic long-term plasticity. NEUROCHEM J+ 2014. [DOI: 10.1134/s181971241403009x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
28
|
Masvekar RR, El-Hage N, Hauser KF, Knapp PE. Morphine enhances HIV-1SF162-mediated neuron death and delays recovery of injured neurites. PLoS One 2014; 9:e100196. [PMID: 24949623 PMCID: PMC4064991 DOI: 10.1371/journal.pone.0100196] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 05/22/2014] [Indexed: 11/19/2022] Open
Abstract
HIV-1 enters the CNS soon after initial systemic infection; within the CNS parenchyma infected and/or activated perivascular macrophages, microglia and astrocytes release viral and cellular toxins that drive secondary toxicity in neurons and other cell types. Our previous work has largely modeled HIV-neuropathology using the individual viral proteins Tat or gp120, with murine striatal neurons as targets. To model disease processes more closely, the current study uses supernatant from HIV-1-infected cells. Supernatant from HIV-1SF162-infected differentiated-U937 cells (HIV+sup) was collected and p24 level was measured by ELISA to assess the infection. Injection drug abuse is a significant risk factor for HIV-infection, and opiate drug abusers show increased HIV-neuropathology, even with anti-retroviral treatments. We therefore assessed HIV+sup effects on neuronal survival and neurite growth/pruning with or without concurrent exposure to morphine, an opiate that preferentially acts through µ-opioid receptors. Effects of HIV+sup ± morphine were assessed on neuronal populations, and also by time-lapse imaging of individual cells. HIV+sup caused dose-dependent toxicity over a range of p24 levels (10–500 pg/ml). Significant interactions occurred with morphine at lower p24 levels (10 and 25 pg/ml), and GSK3β was implicated as a point of convergence. In the presence of glia, selective neurotoxic measures were significantly enhanced and interactions with morphine were also augmented, perhaps related to a decreased level of BDNF. Importantly, the arrest of neurite growth that occurred with exposure to HIV+sup was reversible unless neurons were continuously exposed to morphine. Thus, while reducing HIV-infection levels may be protective, ongoing exposure to opiates may limit recovery. Opiate interactions observed in this HIV-infective environment were similar, though not entirely concordant, with Tat/gp120 interactions reported previously, suggesting unique interactions with virions or other viral or cellular proteins released by infected and/or activated cells.
Collapse
Affiliation(s)
- Ruturaj R. Masvekar
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Nazira El-Hage
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Kurt F. Hauser
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Pamela E. Knapp
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, Virginia, United States of America
- * E-mail:
| |
Collapse
|
29
|
Milanović D, Pešić V, Popić J, Tanić N, Kanazir S, Jevtović-Todorović V, Ruždijić S. Propofol anesthesia induces proapoptotic tumor necrosis factor-α and pro-nerve growth factor signaling and prosurvival Akt and XIAP expression in neonatal rat brain. J Neurosci Res 2014; 92:1362-73. [PMID: 24827783 DOI: 10.1002/jnr.23409] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 02/27/2014] [Accepted: 04/08/2014] [Indexed: 11/12/2022]
Abstract
Previously we observed that prolonged exposure to propofol anesthesia causes caspase-3- and calpain-mediated neuronal death in the developing brain. The present study examines the effects of propofol anesthesia on the expression of tumor necrosis factor-α (TNFα), pro-nerve growth factor (NGF), and their receptors in the cortex and the thalamus. We also investigated how propofol influences the expression of Akt and X-linked inhibitor of apoptosis (XIAP) expression, proteins that promote prosurvival pathways. Seven-day-old rats (P7) were exposed to propofol anesthesia lasting 2, 4, or 6 hr and killed 0, 4, 16, or 24 hr after anesthesia termination. The relative levels of mRNA and protein expression were estimated by RT-PCR and Western blot analysis, respectively. The treatments caused marked activation of TNFα and its receptor TNFR-1 and pro-NGF and p75(NTR) receptor expression. In parallel with the induction of these prodeath signals, we established that propofol anesthesia promotes increased expression of the prosurvival molecules pAkt and XIAP during the 24-hr postanesthesia period. These results show that different brain structures respond to propofol anesthesia with a time- and duration of exposure-dependent increase in proapoptotic signaling and with concomitant increases in activities of prosurvival proteins. We hypothesized that the fine balance between these opposing processes sustains homeostasis in the immature rat brain and prevents unnecessary damage after exposure to an injurious stimulus. The existence of this highly regulated process provides a time frame for potential therapeutic intervention directed toward suppressing the deleterious component of propofol anesthesia.
Collapse
Affiliation(s)
- Desanka Milanović
- Department of Neurobiology, Institute for Biological Research, University of Belgrade, Belgrade, Republic of Serbia
| | | | | | | | | | | | | |
Collapse
|
30
|
Wang JY, Chen F, Fu XQ, Ding CS, Zhou L, Zhang XH, Luo ZG. Caspase-3 cleavage of dishevelled induces elimination of postsynaptic structures. Dev Cell 2014; 28:670-84. [PMID: 24631402 DOI: 10.1016/j.devcel.2014.02.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2013] [Revised: 12/17/2013] [Accepted: 02/13/2014] [Indexed: 11/19/2022]
Abstract
During the development of vertebrate neuromuscular junction (NMJ), agrin stabilizes, whereas acetylcholine (ACh) destabilizes AChR clusters, leading to the refinement of synaptic connections. The intracellular mechanism underlying this counteractive interaction remains elusive. Here, we show that caspase-3, the effector protease involved in apoptosis, mediates elimination of AChR clusters. We found that caspase-3 was activated by cholinergic stimulation of cultured muscle cells without inducing cell apoptosis and that this activation was prevented by agrin. Interestingly, inhibition of caspase-3 attenuated ACh agonist-induced dispersion of AChR clusters. Furthermore, we identified Dishevelled1 (Dvl1), a Wnt signaling protein involved in AChR clustering, as the substrate of caspase-3. Blocking Dvl1 cleavage prevented induced dispersion of AChR clusters. Finally, inhibition or genetic ablation of caspase-3 or expression of a caspase-3-resistant form of Dvl1 caused stabilization of aneural AChR clusters. Thus, caspase-3 plays an important role in the elimination of postsynaptic structures during the development of NMJs.
Collapse
MESH Headings
- Acetylcholine/metabolism
- Adaptor Proteins, Signal Transducing/antagonists & inhibitors
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Agrin/physiology
- Animals
- Caspase 3/metabolism
- Cells, Cultured
- Dishevelled Proteins
- Electrophysiology
- Embryo, Mammalian/cytology
- Embryo, Mammalian/metabolism
- Image Processing, Computer-Assisted
- Immunoenzyme Techniques
- Mice
- Mice, Knockout
- Motor Neurons/cytology
- Motor Neurons/metabolism
- Muscle, Skeletal/cytology
- Muscle, Skeletal/metabolism
- Neuromuscular Junction/physiology
- Phosphoproteins/antagonists & inhibitors
- Phosphoproteins/genetics
- Phosphoproteins/metabolism
- RNA, Small Interfering/genetics
- Rats
- Rats, Sprague-Dawley
- Receptors, Cholinergic/metabolism
- Signal Transduction
- Synaptic Potentials/physiology
- Synaptic Transmission
Collapse
Affiliation(s)
- Jin-Yuan Wang
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Graduate School, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Fei Chen
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Xiu-Qing Fu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Graduate School, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Chuang-Shi Ding
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; School of Life Science and Technology, ShanghaiTech University, 319 Yueyang Road, Shanghai 200031, China
| | - Li Zhou
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Graduate School, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Xiao-Hui Zhang
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Zhen-Ge Luo
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Graduate School, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; School of Life Science and Technology, ShanghaiTech University, 319 Yueyang Road, Shanghai 200031, China.
| |
Collapse
|
31
|
Kudryashova IV. Molecular mechanisms of short-term plasticity as a basis of frequency coding: The role of proteolytic systems. NEUROCHEM J+ 2014. [DOI: 10.1134/s1819712414010097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
32
|
Gassó P, Mas S, Molina O, Lafuente A, Bernardo M, Parellada E. Increased susceptibility to apoptosis in cultured fibroblasts from antipsychotic-naïve first-episode schizophrenia patients. J Psychiatr Res 2014; 48:94-101. [PMID: 24128664 DOI: 10.1016/j.jpsychires.2013.09.017] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 09/12/2013] [Accepted: 09/30/2013] [Indexed: 12/19/2022]
Abstract
Altered apoptosis has been proposed as a potential mechanism involved in the abnormal neurodevelopment and neurodegenerative processes associated with schizophrenia. The aim of this study was to investigate in primary fibroblast cultures whether antipsychotic-naïve patients with first-episode schizophrenia have greater apoptotic susceptibility than healthy controls. Cell growth, cell viability and various apoptotic hallmarks (caspase-3 activity, translocation of phosphatidylserine, chromatin condensation and gene expression of AKT1, BAX, BCL2, CASP3, GSK3B and P53) were measured in fibroblast cultures obtained from skin biopsies of patients (n = 11) and healthy controls (n = 8), both in basal conditions and after inducing apoptosis with staurosporine. Compared to controls, cultured fibroblasts from patients showed higher caspase-3 activity and lower BCL2 expression. When exposed to staurosporine, fibroblasts from patients also showed higher caspase-3 activity; a higher percentage of cells with translocated phosphatidylserine and condensed chromatin; and higher p53 expression compared to fibroblasts from controls. No differences in cell viability or cell growth were detected. These results strongly support the hypothesis that first-episode schizophrenia patients may have increased susceptibility to apoptosis, which may be involved in the onset and progression of the disease.
Collapse
Affiliation(s)
- Patricia Gassó
- Dept. Pathological Anatomy, Pharmacology and Microbiology, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | | | | | | | | | | |
Collapse
|
33
|
Caspase 3 involves in neuroplasticity, microglial activation and neurogenesis in the mice hippocampus after intracerebral injection of kainic acid. J Biomed Sci 2013; 20:90. [PMID: 24313976 PMCID: PMC4028745 DOI: 10.1186/1423-0127-20-90] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 12/02/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The roles of caspase 3 on the kainic acid-mediated neurodegeneration, dendritic plasticity alteration, neurogenesis, microglial activation and gliosis are not fully understood. Here, we investigate hippocampal changes using a mouse model that receive a single kainic acid-intracerebral ventricle injection. The effects of caspase 3 inhibition on these changes were detected during a period of 1 to 7 days post kainic acid injection. RESULT Neurodegeneration was assessed by Fluoro-Jade B staining and neuronal nuclei protein (NeuN) immunostaining. Neurogenesis, gliosis, neuritic plasticity alteration and caspase 3 activation were examined using immunohistochemistry. Dendritic plasticity, cleavvage-dependent activation of calcineurin A and glial fibrillary acidic protein cleavage were analyzed by immunoblotting. We found that kainic acid not only induced neurodegeneration but also arouse several caspase 3-mediated molecular and cellular changes including dendritic plasticity, neurogenesis, and gliosis. The acute caspase 3 activation occurred in pyramidal neurons as well as in hilar interneurons. The delayed caspase 3 activation occurred in astrocytes. The co-injection of caspase 3 inhibitor did not rescue kainic acid-mediated neurodegeneration but seriously and reversibly disturb the structural integrity of axon and dendrite. The kainic acid-induced events include microglia activation, the proliferation of radial glial cells, neurogenesis, and calcineurin A cleavage were significantly inhibited by the co-injection of caspase 3 inhibitor, suggesting the direct involvement of caspase 3 in these events. Alternatively, the kainic acid-mediated astrogliosis is not caspase 3-dependent, although caspase 3 cleavage of glial fibrillary acidic protein occurred. CONCLUSIONS Our results provide the first direct evidence of a causal role of caspase 3 activation in the cellular changes during kainic acid-mediated excitotoxicity. These findings may highlight novel pharmacological strategies to arrest disease progression and control seizures that are refractory to classical anticonvulsant treatment.
Collapse
|
34
|
Sheng M, Ertürk A. Long-term depression: a cell biological view. Philos Trans R Soc Lond B Biol Sci 2013; 369:20130138. [PMID: 24298141 DOI: 10.1098/rstb.2013.0138] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Recent studies of the molecular mechanisms of long-term depression (LTD) suggest a crucial role for the signalling pathways of apoptosis (programmed cell death) in the weakening and elimination of synapses and dendritic spines. With this backdrop, we suggest that LTD can be considered as the electrophysiological aspect of a larger cell biological programme of synapse involution, which uses localized apoptotic mechanisms to sculpt synapses and circuits without causing cell death.
Collapse
Affiliation(s)
- Morgan Sheng
- Department of Neuroscience, Genentech, Inc., , 1 DNA Way, South San Francisco, CA 94080, USA
| | | |
Collapse
|
35
|
Federighi G, Traina G, Macchi M, Ciampini C, Bernardi R, Baldi E, Bucherelli C, Brunelli M, Scuri R. Modulation of gene expression in contextual fear conditioning in the rat. PLoS One 2013; 8:e80037. [PMID: 24278235 PMCID: PMC3837011 DOI: 10.1371/journal.pone.0080037] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 09/27/2013] [Indexed: 01/30/2023] Open
Abstract
In contextual fear conditioning (CFC) a single training leads to long-term memory of context-aversive electrical foot-shocks association. Mid-temporal regions of the brain of trained and naive rats were obtained 2 days after conditioning and screened by two-directional suppression subtractive hybridization. A pool of differentially expressed genes was identified and some of them were randomly selected and confirmed with qRT-PCR assay. These transcripts showed high homology for rat gene sequences coding for proteins involved in different cellular processes. The expression of the selected transcripts was also tested in rats which had freely explored the experimental apparatus (exploration) and in rats to which the same number of aversive shocks had been administered in the same apparatus, but temporally compressed so as to make the association between painful stimuli and the apparatus difficult (shock-only). Some genes resulted differentially expressed only in the rats subjected to CFC, others only in exploration or shock-only rats, whereas the gene coding for translocase of outer mitochondrial membrane 20 protein and nardilysin were differentially expressed in both CFC and exploration rats. For example, the expression of stathmin 1 whose transcripts resulted up regulated was also tested to evaluate the transduction and protein localization after conditioning.
Collapse
Affiliation(s)
- Giuseppe Federighi
- Dipartimento di Ricerca Traslazionale e Delle Nuove Tecnologie in Medicina e Chirurgia, Unità di Fisiologia, Università di Pisa, Pisa, Italy
| | - Giovanna Traina
- Dipartimento di Scienze Economico-Estimative e degli Alimenti, Sezione di Chimica Bromatologica, Biochimica, Fisiologia e Nutrizione, Università degli Studi di Perugia, Perugia, Italy
| | - Monica Macchi
- Dipartimento di Biologia, Università di Pisa, Pisa, Italy
| | - Cristina Ciampini
- Dipartimento di Ricerca Traslazionale e Delle Nuove Tecnologie in Medicina e Chirurgia, Unità di Fisiologia, Università di Pisa, Pisa, Italy
| | - Rodolfo Bernardi
- Dipartimento di Scienze Agrarie, Genetica Alimentari e Agro-Ambientali, Università di Pisa, Pisa, Italy
| | - Elisabetta Baldi
- Dipartimento di Scienze Fisiologiche, Università di Firenze, Firenze, Italy
| | - Corrado Bucherelli
- Dipartimento di Scienze Fisiologiche, Università di Firenze, Firenze, Italy
| | | | - Rossana Scuri
- Dipartimento di Ricerca Traslazionale e Delle Nuove Tecnologie in Medicina e Chirurgia, Unità di Fisiologia, Università di Pisa, Pisa, Italy
| |
Collapse
|
36
|
de Graaf EL, Vermeij WP, de Waard MC, Rijksen Y, van der Pluijm I, Hoogenraad CC, Hoeijmakers JHJ, Altelaar AFM, Heck AJR. Spatio-temporal analysis of molecular determinants of neuronal degeneration in the aging mouse cerebellum. Mol Cell Proteomics 2013; 12:1350-62. [PMID: 23399551 DOI: 10.1074/mcp.m112.024950] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The accumulation of cellular damage, including DNA damage, is hypothesized to contribute to aging-related neurodegenerative changes. DNA excision repair cross-complementing group 1 (Ercc1) knock-out mice represent an accepted model of neuronal aging, showing gradual neurodegenerative changes, including loss of synaptic contacts and cell body shrinkage. Here, we used the Purkinje cell-specific Ercc1 DNA-repair knock-out mouse model to study aging in the mouse cerebellum. We performed an in-depth quantitative proteomics analysis, using stable isotope dimethyl labeling, to decipher changes in protein expression between the early (8 weeks), intermediate (16 weeks), and late (26 weeks) stages of the phenotypically aging Ercc1 knock-out and healthy littermate control mice. The expression of over 5,200 proteins from the cerebellum was compared quantitatively, whereby 79 proteins (i.e. 1.5%) were found to be substantially regulated during aging. Nearly all of these molecular markers of the early aging onset belonged to a strongly interconnected network involved in excitatory synaptic signaling. Using immunohistological staining, we obtained temporal and spatial profiles of these markers confirming not only the proteomics data but in addition revealed how the change in protein expression correlates to synaptic changes in the cerebellum. In summary, this study provides a highly comprehensive spatial and temporal view of the dynamic changes in the cerebellum and Purkinje cell signaling in particular, indicating that synapse signaling is one of the first processes to be affected in this premature aging model, leading to neuron morphological changes, neuron degeneration, inflammation, and ultimately behavior disorders.
Collapse
Affiliation(s)
- Erik L de Graaf
- Biomolecular Mass Spectrometry and Proteomics Group, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Lodovichi C, Belluscio L. Odorant receptors in the formation of the olfactory bulb circuitry. Physiology (Bethesda) 2012; 27:200-12. [PMID: 22875451 DOI: 10.1152/physiol.00015.2012] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
In mammals, smell is mediated by odorant receptors expressed by sensory neurons in the nose. These specialized receptors are found both on olfactory sensory neurons' cilia and axon terminals. Although the primary function of ciliary odorant receptors is to detect odorants, their axonal role remains unclear but is thought to involve axon guidance. This review discusses findings that show axonal odorant receptors are indeed functional and capable of modulating neural connectivity.
Collapse
Affiliation(s)
- Claudia Lodovichi
- Venetian Institute of Molecular Medicine, and Institute of Neuroscience-CNR, Padua, Italy
| | | |
Collapse
|
38
|
D'Amelio M, Sheng M, Cecconi F. Caspase-3 in the central nervous system: beyond apoptosis. Trends Neurosci 2012; 35:700-9. [PMID: 22796265 DOI: 10.1016/j.tins.2012.06.004] [Citation(s) in RCA: 178] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 06/14/2012] [Accepted: 06/15/2012] [Indexed: 01/06/2023]
Abstract
Caspase-3 has been identified as a key mediator of neuronal programmed cell death. This protease plays a central role in the developing nervous system and its activation is observed early in neural tube formation and persists during postnatal differentiation of the neural network. Caspase-3 activation, a crucial event of neuronal cell death program, is also a feature of many chronic neurodegenerative diseases. This traditional apoptotic function of caspase-3 is challenged by recent studies that reveal new cell death-independent roles for mitochondrial-activated caspase-3 in neurite pruning and synaptic plasticity. These findings underscore the need for further research into the mechanism of action and functions of caspase-3 that may prove useful in the development of novel pharmacological treatments for a diverse range of neurological disorders.
Collapse
Affiliation(s)
- Marcello D'Amelio
- Istituto di Ricovero e Cura a Carattere Scientifico, S. Lucia Foundation, via del Fosso di Fiorano 65, 00143 Rome, Italy.
| | | | | |
Collapse
|
39
|
De Chiara G, Marcocci ME, Sgarbanti R, Civitelli L, Ripoli C, Piacentini R, Garaci E, Grassi C, Palamara AT. Infectious agents and neurodegeneration. Mol Neurobiol 2012; 46:614-38. [PMID: 22899188 PMCID: PMC3496540 DOI: 10.1007/s12035-012-8320-7] [Citation(s) in RCA: 161] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 07/31/2012] [Indexed: 12/19/2022]
Abstract
A growing body of epidemiologic and experimental data point to chronic bacterial and viral infections as possible risk factors for neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease and amyotrophic lateral sclerosis. Infections of the central nervous system, especially those characterized by a chronic progressive course, may produce multiple damage in infected and neighbouring cells. The activation of inflammatory processes and host immune responses cause chronic damage resulting in alterations of neuronal function and viability, but different pathogens can also directly trigger neurotoxic pathways. Indeed, viral and microbial agents have been reported to produce molecular hallmarks of neurodegeneration, such as the production and deposit of misfolded protein aggregates, oxidative stress, deficient autophagic processes, synaptopathies and neuronal death. These effects may act in synergy with other recognized risk factors, such as aging, concomitant metabolic diseases and the host’s specific genetic signature. This review will focus on the contribution given to neurodegeneration by herpes simplex type-1, human immunodeficiency and influenza viruses, and by Chlamydia pneumoniae.
Collapse
Affiliation(s)
- Giovanna De Chiara
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Hedskog L, Zhang S, Ankarcrona M. Strategic role for mitochondria in Alzheimer's disease and cancer. Antioxid Redox Signal 2012; 16:1476-91. [PMID: 21902456 DOI: 10.1089/ars.2011.4259] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
SIGNIFICANCE Detailed knowledge about cell death and cell survival mechanisms and how these pathways are impaired in neurodegenerative disorders and cancer forms the basis for future drug development for these diseases that affect millions of people around the world. RECENT ADVANCES In neurodegenerative disorders such as Alzheimer's disease (AD), cell death pathways are inappropriately activated, resulting in neuronal cell death. In contrast, cancer cells develop resistance to apoptosis by regulating anti-apoptotic proteins signaling via mitochondria. Mounting evidence shows that mitochondrial function is central in both cancer and AD. Cancer cells typically shut down oxidative phosphorylation (OXPHOS) in mitochondria and switch to glycolysis for ATP production, making them resistant to hypoxia. In AD, for example, amyloid-β peptide (Aβ) and reactive oxygen species impair mitochondrial function. Neurons therefore also switch to glycolysis to maintain ATP production and to produce molecules involved in antioxidant metabolism in an attempt to survive. CRITICAL ISSUES One critical difference between cancer cells and neurons is that cancer cells can survive without OXPHOS, while neurons are dependent on OXPHOS for long-term survival. FUTURE DIRECTIONS This review will focus on these abnormalities of mitochondrial function shared in AD and cancer and discuss the potential mechanisms underlying links that may be key steps in the development of therapeutic strategies.
Collapse
Affiliation(s)
- Louise Hedskog
- Department of Neurobiology, Care Sciences and Society (NVS), KI-Alzheimer Disease Research Center, Karolinska Institutet, Stockholm, Sweden
| | | | | |
Collapse
|
41
|
Guilarte TR, Opler M, Pletnikov M. Is lead exposure in early life an environmental risk factor for Schizophrenia? Neurobiological connections and testable hypotheses. Neurotoxicology 2012; 33:560-74. [PMID: 22178136 PMCID: PMC3647679 DOI: 10.1016/j.neuro.2011.11.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Revised: 11/30/2011] [Accepted: 11/30/2011] [Indexed: 02/06/2023]
Abstract
Schizophrenia is a devastating neuropsychiatric disorder of unknown etiology. There is general agreement in the scientific community that schizophrenia is a disorder of neurodevelopmental origin in which both genes and environmental factors come together to produce a schizophrenia phenotype later in life. The challenging questions have been which genes and what environmental factors? Although there is evidence that different chromosome loci and several genes impart susceptibility for schizophrenia; and epidemiological studies point to broad aspects of the environment, only recently there has been an interest in studying gene × environment interactions. Recent evidence of a potential association between prenatal lead (Pb(2+)) exposure and schizophrenia precipitated the search for plausible neurobiological connections. The most promising connection is that in schizophrenia and in developmental Pb(2+) exposure there is strong evidence for hypoactivity of the N-methyl-d-aspartate (NMDA) subtype of excitatory amino acid receptors as an underlying neurobiological mechanism in both conditions. A hypofunction of the NMDA receptor (NMDAR) complex during critical periods of development may alter neurobiological processes that are essential for brain growth and wiring, synaptic plasticity and cognitive and behavioral outcomes associated with schizophrenia. We also describe on-going proof of concept gene-environment interaction studies of early life Pb(2+) exposure in mice expressing the human mutant form of the disrupted in schizophrenia 1 (DISC-1) gene, a gene that is strongly associated with schizophrenia and allied mental disorders.
Collapse
Affiliation(s)
- Tomás R Guilarte
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY 10032, United States.
| | | | | |
Collapse
|
42
|
Popic J, Pesic V, Milanovic D, Todorovic S, Kanazir S, Jevtovic-Todorovic V, Ruzdijic S. Propofol-induced changes in neurotrophic signaling in the developing nervous system in vivo. PLoS One 2012; 7:e34396. [PMID: 22496799 PMCID: PMC3319585 DOI: 10.1371/journal.pone.0034396] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 02/27/2012] [Indexed: 01/19/2023] Open
Abstract
Several studies have revealed a role for neurotrophins in anesthesia-induced neurotoxicity in the developing brain. In this study we monitored the spatial and temporal expression of neurotrophic signaling molecules in the brain of 14-day-old (PND14) Wistar rats after the application of a single propofol dose (25 mg/kg i.p). The structures of interest were the cortex and thalamus as the primary areas of anesthetic actions. Changes of the protein levels of the brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF), their activated receptors tropomyosin-related kinase (TrkA and TrkB) and downstream kinases Akt and the extracellular signal regulated kinase (ERK) were assessed by Western immunoblot analysis at different time points during the first 24 h after the treatment, as well as the expression of cleaved caspase-3 fragment. Fluoro-Jade B staining was used to follow the appearance of degenerating neurons. The obtained results show that the treatment caused marked alterations in levels of the examined neurotrophins, their receptors and downstream effector kinases. However, these changes were not associated with increased neurodegeneration in either the cortex or the thalamus. These results indicate that in the brain of PND14 rats, the interaction between Akt/ERK signaling might be one of important part of endogenous defense mechanisms, which the developing brain utilizes to protect itself from potential anesthesia-induced damage. Elucidation of the underlying molecular mechanisms will improve our understanding of the age-dependent component of anesthesia-induced neurotoxicity.
Collapse
Affiliation(s)
- Jelena Popic
- Department of Neurobiology, Institute for Biological Research, University of Belgrade, Belgrade, Serbia
| | - Vesna Pesic
- Department of Neurobiology, Institute for Biological Research, University of Belgrade, Belgrade, Serbia
| | - Desanka Milanovic
- Department of Neurobiology, Institute for Biological Research, University of Belgrade, Belgrade, Serbia
- Department of Anesthesiology, University of Virginia Health System, Charlottesville, Virginia, United States of America
| | - Smilja Todorovic
- Department of Neurobiology, Institute for Biological Research, University of Belgrade, Belgrade, Serbia
| | - Selma Kanazir
- Department of Neurobiology, Institute for Biological Research, University of Belgrade, Belgrade, Serbia
| | - Vesna Jevtovic-Todorovic
- Department of Anesthesiology, University of Virginia Health System, Charlottesville, Virginia, United States of America
| | - Sabera Ruzdijic
- Department of Neurobiology, Institute for Biological Research, University of Belgrade, Belgrade, Serbia
- * E-mail:
| |
Collapse
|
43
|
Rőszer T, Bánfalvi G. FMRFamide-related peptides: anti-opiate transmitters acting in apoptosis. Peptides 2012; 34:177-85. [PMID: 21524675 DOI: 10.1016/j.peptides.2011.04.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2011] [Revised: 04/04/2011] [Accepted: 04/07/2011] [Indexed: 01/10/2023]
Abstract
Members of the FMRFamide-related peptide (FaRP) family are neurotransmitters, hormone-like substances and tumor suppressor peptides. In mammals, FaRPs are considered as anti-opiate peptides due to their ability to inhibit opioid signaling. Some FaRPs are asserted to attenuate opiate tolerance. A recently developed chimeric FaRP (Met-enkephalin-FMRFa) mimics the analgesic effects of opiates without the development of opiate-dependence, displaying a future therapeutical potential in pain reduction. In this review we support the notion, that opiates and representative members of the FaRP family show overlapping effects on apoptosis. Binding of FaRPs to opioid receptors or to their own receptors (G-protein linked membrane receptors and acid-sensing ion channels) evokes or suppresses cell death, in a cell- and receptor-type manner. With the dramatically increasing incidence of opiate abuse and addiction, understanding of opioid-induced cell death, and in this context FaRPs will deserve growing attention.
Collapse
Affiliation(s)
- Tamás Rőszer
- Department of Microbial Biotechnology & Cell Biology, University of Debrecen, Debrecen, Hungary.
| | | |
Collapse
|
44
|
Hedskog L, Petersen CAH, Svensson AI, Welander H, Tjernberg LO, Karlström H, Ankarcrona M. γ-Secretase complexes containing caspase-cleaved presenilin-1 increase intracellular Aβ(42) /Aβ(40) ratio. J Cell Mol Med 2012; 15:2150-63. [PMID: 21054783 PMCID: PMC4394225 DOI: 10.1111/j.1582-4934.2010.01208.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Markers for caspase activation and apoptosis have been shown in brains of Alzheimer's disease (AD) patients and AD-mouse models. In neurons, caspase activation is associated with elevated amyloid β-peptide (Aβ) production. Caspases cleave numerous substrates including presenilin-1 (PS1). The cleavage takes place in the large cytosolic loop of PS1-C-terminal fragment (PS1CTF), generating a truncated PS1CTF lacking half of the loop domain (caspCTF). The loop has been shown to possess important regulatory functions with regard to Aβ(40) and Aβ(42) production. Previously, we have demonstrated that γ-secretase complexes are active during apoptosis regardless of caspase cleavage in the PS1CTF-loop. Here, a PS1/PS2-knockout mouse blastocyst-derived cell line was used to establish stable or transient cell lines expressing either caspCTF or full-length CTF (wtCTF). We show that caspCTF restores γ-secretase activity and forms active γ-secretase complexes together with Nicastrin, Pen-2, Aph-1 and PS1-N-terminal fragment. Further, caspCTF containing γ-secretase complexes have a sustained capacity to cleave amyloid precursor protein (APP) and Notch, generating APP and Notch intracellular domain, respectively. However, when compared to wtCTF cells, caspCTF cells exhibit increased intracellular production of Aβ(42) accompanied by increased intracellular Aβ(42) /Aβ(40) ratio without changing the Aβ secretion pattern. Similarly, induction of apoptosis in wtCTF cells generate a similar shift in intracellular Aβ pattern with increased Aβ(42) /Aβ(40) ratio. In summary, we show that caspase cleavage of PS1 generates a γ-secretase complex that increases the intracellular Aβ(42) /Aβ(40) ratio. This can have implications for AD pathogenesis and suggests caspase inhibitors as potential therapeutic agents.
Collapse
Affiliation(s)
- Louise Hedskog
- KI-Alzheimer's Disease Research Center, Karolinska Institutet, Department of Neurobiology, Care Sciences and Society, Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
45
|
Keeler BE, Liu G, Siegfried RN, Zhukareva V, Murray M, Houlé JD. Acute and prolonged hindlimb exercise elicits different gene expression in motoneurons than sensory neurons after spinal cord injury. Brain Res 2011; 1438:8-21. [PMID: 22244304 DOI: 10.1016/j.brainres.2011.12.015] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Revised: 12/06/2011] [Accepted: 12/09/2011] [Indexed: 01/31/2023]
Abstract
We examined gene expression in the lumbar spinal cord and the specific response of motoneurons, intermediate gray and proprioceptive sensory neurons after spinal cord injury and exercise of hindlimbs to identify potential molecular processes involved in activity dependent plasticity. Adult female rats received a low thoracic transection and passive cycling exercise for 1 or 4weeks. Gene expression analysis focused on the neurotrophic factors: brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF), neurotrophin-3 (NT-3), neurotrophin-4 (NT-4), and their receptors because of their potential roles in neural plasticity. We also examined expression of genes involved in the cellular response to injury: heat shock proteins (HSP) -27 and -70, glial fibrillary acidic protein (GFAP) and caspases -3, -7, and -9. In lumbar cord samples, injury increased the expression of mRNA for TrkB, all three caspases and the HSPs. Acute and prolonged exercise increased expression of mRNA for the neurotrophic factors BDNF and GDNF, but not their receptors. It also increased HSP expression and decreased caspase-7 expression, with changes in protein levels complimentary to these changes in mRNA expression. Motoneurons and intermediate gray displayed little change in mRNA expression following injury, but acute and prolonged exercise increased levels of mRNA for BDNF, GDNF and NT-4. In large DRG neurons, mRNA for neurotrophic factors and their receptors were largely unaffected by either injury or exercise. However, caspase mRNA expression was increased by injury and decreased by exercise. Our results demonstrate that exercise affects expression of genes involved in plasticity and apoptosis in a cell specific manner and that these change with increased post-injury intervals and/or prolonged periods of exercise.
Collapse
Affiliation(s)
- Benjamin E Keeler
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | |
Collapse
|
46
|
Kudryashova IV. Structural and functional modifications of presynaptic afferents: Do they correlate with learning mechanisms? NEUROCHEM J+ 2011. [DOI: 10.1134/s181971241104009x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
47
|
Abstract
Axon guidance is a crucial part of neural circuit formation. While precise axonal targeting forms the basis of accurate information delivery, the mechanisms that regulate this process are still unclear. Apoptotic signaling molecules have been identified in the axon terminal, but their specific role in axon guidance is not well understood. Here we use the mouse olfactory system as an in vivo model to demonstrate that by modulating Fas-associated factor 1 (FAF1), an apoptosis regulatory molecule, we can rewire axonal projections. Interestingly, FAF1 is highly expressed in the developing mouse olfactory system, but its expression is downregulated postnatally. Using a tetracycline-inducible promoter Tet-Off system, we generated transgenic mice in which FAF1 is specifically expressed in immature olfactory sensory neurons (OSNs) and show that overexpression of FAF1 not only misroutes OSN axons to deep layers of the olfactory bulb but also leads to widespread disruption of the glomerular layer. In addition, we also demonstrate that the specific convergence of P2 receptor OSN axons is completely distorted in the FAF1 mice. Strikingly, all of the mutant phenotypes can be recovered by shutting down FAF1 expression through the administration of doxycycline. Together, our study provides clear in vivo evidence that an apoptotic molecule can indeed regulate axon targeting and that OSNs can restore their organization even after broad disruption.
Collapse
|
48
|
Yakovlev AA, Gulyaeva NV. Pleiotropic functions of brain proteinases: Methodological considerations and search for caspase substrates. BIOCHEMISTRY (MOSCOW) 2011; 76:1079-86. [DOI: 10.1134/s0006297911100014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
49
|
Abstract
In the past few years, the development of pharmaceutical agents that enhance the physiological effects of glucagon-like peptide-1 (GLP-1), either through GLP-1 receptor agonism (GLP-1 agonists) or by inhibiting GLP-1 degradation (dipeptidylpeptidase-4 inhibitors) has broadened the range of treatment options for individuals with type 2 diabetes. It has been recognized for some time that GLP-1 also has extra-pancreatic effects, notably targeting the brain, where it regulates appetite and satiety, as well as peripheral functions highly controlled by the autonomic nervous system, such as gastric emptying. Furthermore, data are beginning to emerge that indicate a potential role for GLP-1 in neuroprotection. The increased risk of Alzheimer's disease, Parkinson's disease and stroke in people with type 2 diabetes suggests that shared mechanisms/pathways of cell death, possibly related to insulin dysregulation, may underlie all of these disorders. Although the disease anatomy varies with each disorder, a wide range of genetic and environmental triggers result in activation of similar biochemical pathways in all of them, suggesting a complex network of biochemical events that feed in to a final common path towards cellular dysfunction and death. This article summarizes the evidence for neuronal activity of GLP-1 and examines the limited data that currently exist on the therapeutic potential of GLP-1 in specific neurological and neurodegenerative conditions, namely Alzheimer's disease, Parkinson's disease, Huntingdon's disease, stroke and peripheral sensory neuropathy.
Collapse
Affiliation(s)
- Jens Juul Holst
- Department of Biomedical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, Denmark.
| | | | | |
Collapse
|
50
|
Abstract
Increased Lymphocyte Caspase-3 Activity in Patients with SchizophreniaA growing body of evidence indicates that cortical brain cells of schizophrenic patients are vulnerable to apoptosis. As apoptosis is an important mechanism in organism modeling during development, active since the early phase of intrauterine life, it could be involved in the pathogenesis of schizophrenia. To test this hypothesis, caspase-3 activity was determined in peripheral blood mono nuclear cells from 30 patients with schizophrenia and from 30 age and gender matched healthy subjects by a colorimetric commercially available kit. Consistent with increased susceptibility to apoptosis, caspase-3 activity in lymphocytes of patients with schizophrenia was significantly increased (0.111±0.055 μmol/mg protein, p<0.05) in comparison with those in the matched control group (0.086±0.030 μmol/mg protein). The highest activity was obtained in the group showing almost equally positive and negative symptoms (0.159±0.096 μmol/mg protein) and it was significantly higher (p<0.05) compared to the group with a relative predomination of positive symptoms (0.100±0.029 μmol/mg protein). Caspase-3 activity in patients receiving typical antipsychotic drugs (0.124± 0.071 μmol/mg protein) was not significantly different from that in patients treated with atypical antipsychotics (0.104±0.039 μmol/mg protein). To our knowledge to date, this has been the first demonstration that there is a significant increase in caspase-3 activity, determined in native cells, in patients with schizophrenia, indicating a dysregulated apoptotic mechanism in this disease.
Collapse
|