1
|
Ngamratanapaiboon S, Srikornvit N, Hongthawonsiri P, Pornchokchai K, Wongpitoonmanachai S, Mo J, Pholkla P, Yambangyang P, Duchda P, Lohwacharin J, Ayutthaya WDN. Elucidating of the metabolic impact of risperidone on brain microvascular endothelial cells using untargeted metabolomics-based LC-MS. Toxicol Rep 2024; 13:101691. [PMID: 39104367 PMCID: PMC11299597 DOI: 10.1016/j.toxrep.2024.101691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/01/2024] [Accepted: 07/07/2024] [Indexed: 08/07/2024] Open
Abstract
Risperidone is useful for the treatment of schizophrenia symptoms; however, it also has side effects, and an overdose can be harmful. The metabolic effects of risperidone at high therapeutic doses and its metabolites have not been elucidated. Endogenous cellular metabolites may be comprehensively analyzed using untargeted metabolomics-based liquid chromatography-mass spectrometry (LC-MS), which can reveal changes in cell regulation and metabolic pathways. By identifying the metabolites and pathway changes using a nontargeted metabolomics-based LC-MS approach, we aimed to shed light on the potential toxicological effects of high-dose risperidone on brain microvascular endothelial cells (MVECs) associated with the human blood brain barrier. A total of 42 metabolites were selected as significant putative metabolites of the toxicological response of high-dose risperidone in MVECs. Six highly correlated pathways were identified, including those involving diacylglycerol, fatty acid, ceramide, glycerophospholipid, amino acid, and tricarboxylic acid metabolism. We demonstrated that methods focused on metabolomics are useful for identifying metabolites that may be used to clarify the mechanism of drug-induced toxicity.
Collapse
Affiliation(s)
- Surachai Ngamratanapaiboon
- Division of Pharmacology, Department of Basic Medical Sciences, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok 10300, Thailand
| | - Napatarin Srikornvit
- Medical Student in Doctor of Medicine Programme, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok 10300, Thailand
| | - Patipol Hongthawonsiri
- Medical Student in Doctor of Medicine Programme, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok 10300, Thailand
| | - Krittaboon Pornchokchai
- Medical Student in Doctor of Medicine Programme, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok 10300, Thailand
| | - Siriphattarinya Wongpitoonmanachai
- Medical Student in Doctor of Medicine Programme, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok 10300, Thailand
| | - Jiajun Mo
- Medical Student in Doctor of Medicine Programme, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok 10300, Thailand
| | - Petchlada Pholkla
- Medical Student in Doctor of Medicine Programme, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok 10300, Thailand
| | - Pracha Yambangyang
- Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Salaya, Nakhon Pathom 73170, Thailand
| | - Phichanan Duchda
- Department of Environmental Engineering, Faculty of Engineering, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand
- Professor Aroon Sorathesn Center of Excellence in Environmental Engineering, Department of Environmental Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Jenyuk Lohwacharin
- Department of Environmental Engineering, Faculty of Engineering, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand
- Professor Aroon Sorathesn Center of Excellence in Environmental Engineering, Department of Environmental Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Watcharaporn Devakul Na Ayutthaya
- Division of Pharmacology, Department of Basic Medical Sciences, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok 10300, Thailand
| |
Collapse
|
2
|
Alenazi B, Al Doghaither HA, Al-Ghafari AB, Elmorsy EM. Risperidone-induced bioenergetic disruption in the isolated human peripheral blood monocytes. Toxicol In Vitro 2024; 101:105936. [PMID: 39237056 DOI: 10.1016/j.tiv.2024.105936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 08/23/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024]
Abstract
Risperidone (RIS) is a widely used antipsychotic drug with reported alteration in immune response. The current study investigated mitochondrial disruption as the underlying mechanism of RIS-induced immunotoxicity in isolated human peripheral blood monocytes (hPBM). RIS was cytotoxic to hPBM in exposure duration and concentration-dependent patterns. Functionally, RIS was shown to increase the release of IL-6, TNF-α, and IL-8 with a decrease in test particle phagocytosis in concertation and exposure time-based patterns. It was found that RIS decreased ATP production in isolated monocytes' mitochondria, with an estimated EC50 of around 70 μM after 24 h with parallel inhibition of mitochondrial complexes I and III activities and decreased mitochondrial membrane potential and oxygen consumption rates with increased lactate production from by the treated cells in comparison to controls. Structurally, RIS in 100 μM concentration significantly increased the mitochondrial membrane fluidity with significant increase in increased unsaturated/saturated fatty acids ratios of the mitochondrial membranes of the treated cells. Interestingly, water-soluble CoQ10 formulation significantly decreased the cytotoxic effect of RIS and improved the phagocytic activity of RIS-treated cells. To conclude, the current data suggests mitochondrial disruption as the underlying mechanism of RIS-induced immunotoxicity with shown protective effect of water-soluble CoQ10 formulation.
Collapse
Affiliation(s)
- Bandar Alenazi
- Pharmacology Department, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia
| | - Huda A Al Doghaither
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ayat B Al-Ghafari
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ekramy M Elmorsy
- Pathology Department, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia.
| |
Collapse
|
3
|
Elmorsy EM, Al-Ghafari AB, Al Doghaither HA. Fucoxanthin alleviates the cytotoxic effects of cadmium and lead on a human osteoblast cell line. Toxicol Res (Camb) 2024; 13:tfae218. [PMID: 39712643 PMCID: PMC11655842 DOI: 10.1093/toxres/tfae218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/23/2024] [Accepted: 12/09/2024] [Indexed: 12/24/2024] Open
Abstract
OBJECTIVE Cadmium (Cd) and lead (Pb) are non-biodegradable heavy metals (HMs) that persistently contaminate ecosystems and accumulate in bones, where they exert harmful effects. This study aimed to investigate the protective effect of fucoxanthin (FX) against the chemical toxicity induced by Cd and Pb in human bone osteoblasts in vitro, using various biochemical and molecular assays. METHODS The effect of metals and FX on osteoblasts viability was assayed by MTT, then the effect of Pb, Cd, and FX on the cells' mitochondrial parameters was studied via assays for ATP, mitochondrial membrane potential (MMP), mitochondrial complexes, and lactate production. Also, the effect of metals on oxidative stress was assessed by reactive oxygen species, lipid peroxidation and antioxidant enzymes assays. Also the effect of FX and metals on apoptosis caspases and related genes was assessed. RESULTS When Cd and Pb were added to human osteoblast cultures at concentrations ranging from 1-20 μM for 72 h, they significantly reduced osteoblast viability in a time and concentration-dependent manner. The cytotoxic effect of Cd on osteoblasts was greater than that of Pb, with estimated EC50 of 8 and 12 μM, respectively, after 72 h of exposure. FX (10 and 20 μM) alleviated the cytotoxicity of the metals. Bioenergetics assays, including ATP, MMP, and mitochondrial complexes I and III activities, revealed that HMs at 1 and 10 μM concentrations inhibited cellular bioenergetics after 72 h of exposure. Cd and Pb also increased lipid peroxidation and reactive oxygen species while reducing catalase and superoxide dismutase antioxidant activities and oxidative stress-related genes. This was accompanied by increased caspases -3, -8, and - 9 and Bax/bCl-2 ratio. Co-treatment with FX (10 and 20 μM) mitigated the disruption of bioenergetics, oxidative damage, and apoptosis induced by the metals, showing a concentration-dependent pattern to varying extents. CONCLUSION These findings strongly support the role of FX in managing toxicities induced by environmental pollutants in bones and in addressing bone diseases associated with molecular bases of oxidative stress, apoptosis, and bioenergetic disruption.
Collapse
Affiliation(s)
- Ekramy M Elmorsy
- Pathology Department, Faculty of Medicine, Northern Border University, Arar 91431, Saudi Arabia
- Center for Health Research, Northern Border University, Arar 91431, Saudi Arabia
| | - Ayat B Al-Ghafari
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Huda A Al Doghaither
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
4
|
Demirlek C, Arslan B, Eyuboglu MS, Yalincetin B, Atas F, Cesim E, Demir M, Uzman Ozbek S, Kizilay E, Verim B, Sut E, Baykara B, Kaya M, Akdede BB, Bora E. Retina in Clinical High-Risk and First-Episode Psychosis. Schizophr Bull 2024:sbae189. [PMID: 39488000 DOI: 10.1093/schbul/sbae189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2024]
Abstract
BACKGROUND AND HYPOTHESIS Abnormalities in the retina are observed in psychotic disorders, especially in schizophrenia. STUDY DESIGN Using spectral-domain optical coherence tomography, we investigated structural retinal changes in relatively metabolic risk-free youth with clinical high-risk (CHR, n = 34) and first-episode psychosis (FEP, n = 30) compared with healthy controls (HCs, n = 28). STUDY RESULTS Total retinal macular thickness/volume of the right eye increased in FEP (effect sizes, Cohen's d = 0.69/0.66) and CHR (d = 0.67/0.76) compared with HCs. Total retinal thickness/volume was not significantly different between FEP and CHR. Macular retinal nerve fiber layer (RNFL) thickness/volume of the left eye decreased in FEP compared with HCs (d = -0.75/-0.66). Peripapillary RNFL thickness was not different between groups. The ganglion cell (GCL), inner plexiform (IPL), and inner nuclear (INL) layers thicknesses/volumes of both eyes increased in FEP compared with HCs (d = 0.70-1.03). GCL volumes of both eyes, IPL thickness/volume of the left eye, and INL thickness/volume of both eyes increased in CHR compared with HCs (d = 0.64-1.01). In the macula, while central sector thickness/volume decreased (d = -0.62 to -0.72), superior outer (peri-foveal) sector thickness/volume of both eyes increased (d = 0.81 to 0.86) in FEP compared with HCs. CONCLUSIONS The current findings suggest that distinct regions and layers of the retina may be differentially impacted during the emergence and early phase of psychosis. Consequently, oculomics could play significant roles, not only as a diagnostic tool but also as a mirror reflecting neurobiological changes at axonal and cellular levels.
Collapse
Affiliation(s)
- Cemal Demirlek
- Department of Neurosciences, Institute of Health Sciences, Dokuz Eylul University, Izmir 35340, Turkey
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA 02478, United States
| | - Berat Arslan
- Department of Neurosciences, Institute of Health Sciences, Dokuz Eylul University, Izmir 35340, Turkey
| | - Merve S Eyuboglu
- Department of Neurosciences, Institute of Health Sciences, Dokuz Eylul University, Izmir 35340, Turkey
| | - Berna Yalincetin
- Department of Neurosciences, Institute of Health Sciences, Dokuz Eylul University, Izmir 35340, Turkey
| | - Ferdane Atas
- Department of Ophthalmology, Marmara University, Faculty of Medicine, Istanbul 34854, Turkey
| | - Ezgi Cesim
- Department of Neurosciences, Institute of Health Sciences, Dokuz Eylul University, Izmir 35340, Turkey
| | - Muhammed Demir
- Department of Neurosciences, Institute of Health Sciences, Dokuz Eylul University, Izmir 35340, Turkey
| | - Simge Uzman Ozbek
- Department of Psychiatry, Faculty of Medicine, Dokuz Eylul University, Izmir 35340, Turkey
| | - Elif Kizilay
- Department of Neurosciences, Institute of Health Sciences, Dokuz Eylul University, Izmir 35340, Turkey
| | - Burcu Verim
- Department of Neurosciences, Institute of Health Sciences, Dokuz Eylul University, Izmir 35340, Turkey
| | - Ekin Sut
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Dokuz Eylul University, Izmir 35340, Turkey
| | - Burak Baykara
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Dokuz Eylul University, Izmir 35340, Turkey
| | | | - Berna B Akdede
- Department of Neurosciences, Institute of Health Sciences, Dokuz Eylul University, Izmir 35340, Turkey
- Department of Psychiatry, Faculty of Medicine, Dokuz Eylul University, Izmir 35340, Turkey
| | - Emre Bora
- Department of Neurosciences, Institute of Health Sciences, Dokuz Eylul University, Izmir 35340, Turkey
- Department of Psychiatry, Faculty of Medicine, Dokuz Eylul University, Izmir 35340, Turkey
- Department of Psychiatry, Melbourne Neuropsychiatry Centre, University of Melbourne and Melbourne Health, Victoria 3053, Australia
| |
Collapse
|
5
|
Elmorsy EM, Al-Ghafari AB, Al Doghaither HA, Fawzy MS, Shehata SA. Neurotoxic mechanisms of dexamethasone in SH-SY5Y neuroblastoma cells: Insights into bioenergetics, oxidative stress, and apoptosis. Steroids 2024; 212:109514. [PMID: 39303897 DOI: 10.1016/j.steroids.2024.109514] [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: 06/18/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Despite the known therapeutic uses of dexamethasone (DEX), the specific mechanisms underlying its neurotoxic effects in neuronal cells, particularly in undifferentiated human neuroblastoma (SH-SY5Y) cells, remain inadequately understood. This study aims to elucidate these mechanisms, emphasizing bioenergetics, oxidative stress, and apoptosis, thereby providing novel insights into the cellular vulnerabilities induced by chronic DEX exposure. The findings revealed significant reductions in cell viability, altered membrane integrity with LDH leakage, decreased intracellular ATP production, and the electron transport chain complexes I and III activity inhibition. DEX significantly increased the release of the reactive species and peroxidation of lipids, as well as of Nrf2 expression. At the same time, it simultaneously led to a decline in the activities of the antioxidant catalase and superoxide dismutase enzymes, along with a depletion of glutathione reserves. The apoptosis process was exhibited by a significant elevation of caspases 3 and 8 activities with overexpression of mRNA BAX, inhibition of BCL-2, and a significant upregulation of the BAX/BCL-2 ratio. Assessment of neuronal development genes (GAP43, CAMK2A, CAMK2B, TUBB3, and Wnts) by quantitative PCR assay showed increased expression of CAMK2A, CAMK2B, and Wnt3a with a significant reduction in GAP43 mRNA levels. Collectively, this study proved that DEX was cytotoxic to SH-SY5Y via bioenergetic disruption, mitochondrial dysfunction, oxidative stress, and apoptosis.
Collapse
Affiliation(s)
- Ekramy M Elmorsy
- Pathology Department, Faculty of Medicine, Northern Border University, Arar 91431, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Ayat B Al-Ghafari
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Huda A Al Doghaither
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Manal S Fawzy
- Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar 91431, Saudi Arabia; Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt.
| | - Shaimaa A Shehata
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
6
|
Wakonigg Alonso C, McElhatton F, O'Mahony B, Campbell M, Pollak TA, Stokes PRA. The blood-brain barrier in bipolar disorders: A systematic review. J Affect Disord 2024; 361:434-444. [PMID: 38897301 DOI: 10.1016/j.jad.2024.06.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/13/2024] [Accepted: 06/13/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Bipolar disorders (BD) are chronic, debilitating disorders. The blood-brain barrier (BBB) has been increasingly investigated in BD. This systematic review aimed to assess the available evidence on the relationship between BD and markers of BBB dysfunction. METHODS A systematic search in PubMed, Embase, PsycINFO, CINAHL and Web of Science was run where the primary outcomes were BBB markers such as S100B, albumin ratio, matrix metalloproteinase (MMP), cell adhesion molecule (CAM), and tight junction proteins. Techniques included blood, cerebrospinal fluid (CSF), post-mortem, genetic and imaging methods in BD compared to healthy controls. RESULTS 55 studies were identified, 38 of which found an association between BD and markers of BBB dysfunction. 16/29 studies found increased blood/CSF albumin ratio, S100B, CAMs or MMP levels in BD participants compared to controls. 5/19 post-mortem studies found increased levels of chondroitin sulphate proteoglycans, intercellular CAM, neurexin or claudin-5 mRNA in distinct locations throughout the brain in BD compared to controls. One imaging study identified extensive BBB leakage in 30 % of BD participants, compared to 0 % in controls. LIMITATIONS The diversity in methodologies used in the included studies makes direct comparison of results challenging. Furthermore, imaging methods are the gold standard, but only one study used them. Other markers are only indicative of BBB permeability. CONCLUSIONS This review suggests an association between BD and BBB dysfunction. Further research is needed to provide definite answers considering the existing literature's limitations, and to clarify whether this association provides a pathogenic mechanism, or is an epiphenomenon of BD.
Collapse
Affiliation(s)
- Clara Wakonigg Alonso
- Institute of Psychiatry & Psychology and Neuroscience, King's College London,United Kingdom.
| | - Frances McElhatton
- Institute of Psychiatry & Psychology and Neuroscience, King's College London,United Kingdom
| | - Brian O'Mahony
- Institute of Psychiatry & Psychology and Neuroscience, King's College London,United Kingdom
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland
| | - Thomas A Pollak
- Dept of Psychosis Studies, Institute of Psychiatry & Psychology and Neuroscience, King's College London, United Kingdom; South London and Maudsley NHS Foundation Trust,Bethlem Royal Hospital, Monks Orchard Road, Beckenham, Kent, BR3 3BX, United Kingdom
| | - Paul R A Stokes
- South London and Maudsley NHS Foundation Trust,Bethlem Royal Hospital, Monks Orchard Road, Beckenham, Kent, BR3 3BX, United Kingdom; Centre for Affective Disorders, Department of Psychological Medicine, Institute of Psychiatry & Psychology and Neuroscience, King's College London,United Kingdom
| |
Collapse
|
7
|
Elmorsy EM, Al-Ghafari AB, Al Doghaither HA, Alrowaili MG, Khired ZA, Toraih EA, Fawzy MS, Shehata SA. Vitamin D Alleviates Heavy Metal-Induced Cytotoxic Effects on Human Bone Osteoblasts Via the Induction of Bioenergetic Disruption, Oxidative Stress, and Apoptosis. Biol Trace Elem Res 2024:10.1007/s12011-024-04337-8. [PMID: 39235540 DOI: 10.1007/s12011-024-04337-8] [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: 06/18/2024] [Accepted: 08/02/2024] [Indexed: 09/06/2024]
Abstract
Cadmium (Cd) and lead (Pb) are heavy metals (HMs) that persistently contaminate the ecosystem, and bioaccumulation in bones is a health concern. We used biochemical and molecular assays to assess the cytoprotective effect of vitamin D (VD) on Cd- and Pd-induced chemical toxicity of human bone osteoblasts in vitro. Exposing Cd and Pb to human osteoblast cultures at concentrations of 0.1-1000 µM for 24-72 h significantly reduced osteoblast viability in an exposure time- and concentration-dependent manner. The cytotoxic effect of Cd on osteoblasts was more severe than Pb's, with 72-h exposure estimated half maximal effective concentration (EC50) of 8 and 12 µM, respectively, and VD (1 and 10 nM) alleviated cytotoxicity. Bioenergetics assays of ATP, mitochondrial membrane potential, and mitochondrial complex I and III activity showed that both Cd and Pb (1 and 10 µM) inhibited cellular bioenergetics after 72-h exposure. Cd and Pb increased lipid peroxidation and reactive oxygen species with reduced catalase/superoxide dismutase antioxidant activities and increased activity of caspases -3, -8, and -9. Co-treatment with VD (1 and 10 nM) counteracted bioenergetic disruption, oxidative damage, and apoptosis in a concentration-dependent manner. These findings suggest that VD is effective in managing the toxic effects of environmental pollutants and in treating bone diseases characterized by oxidative stress, apoptosis, and bioenergetic disruption.
Collapse
Affiliation(s)
- Ekramy M Elmorsy
- Pathology Department, Faculty of Medicine, Northern Border University, 91431, Arar, Saudi Arabia.
- Center for Health Research, Northern Border University, Arar, Saudi Arabia.
| | - Ayat B Al-Ghafari
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Huda A Al Doghaither
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Majed Gorayan Alrowaili
- Department of Surgery (Orthopedic Division), Faculty of Medicine, Northern Border University, Arar, Saudi Arabia
| | - Zenat Ahmed Khired
- Department of Surgery, College of Medicine, Jazan University, 45142, Jazan, Saudi Arabia
| | - Eman A Toraih
- Department of Surgery, School of Medicine, Tulane University, New Orleans, LA, 70112, USA.
- Genetics Unit, Department of Histology and Cell Biology, Suez Canal University, Ismailia, 41522, Egypt.
| | - Manal S Fawzy
- Center for Health Research, Northern Border University, Arar, Saudi Arabia
- Department of Biochemistry, Faculty of Medicine, Northern Border University, 73213, Arar, Saudi Arabia
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Shaimaa A Shehata
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| |
Collapse
|
8
|
Campana M, Yakimov V, Moussiopoulou J, Maurus I, Löhrs L, Raabe F, Jäger I, Mortazavi M, Benros ME, Jeppesen R, Meyer Zu Hörste G, Heming M, Giné-Servén E, Labad J, Boix E, Lennox B, Yeeles K, Steiner J, Meyer-Lotz G, Dobrowolny H, Malchow B, Hansen N, Falkai P, Siafis S, Leucht S, Halstead S, Warren N, Siskind D, Strube W, Hasan A, Wagner E. Association of symptom severity and cerebrospinal fluid alterations in recent onset psychosis in schizophrenia-spectrum disorders - An individual patient data meta-analysis. Brain Behav Immun 2024; 119:353-362. [PMID: 38608742 DOI: 10.1016/j.bbi.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/03/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024] Open
Abstract
Neuroinflammation and blood-cerebrospinal fluid barrier (BCB) disruption could be key elements in schizophrenia-spectrum disorderś(SSDs) etiology and symptom modulation. We present the largest two-stage individual patient data (IPD) meta-analysis, investigating the association of BCB disruption and cerebrospinal fluid (CSF) alterations with symptom severity in first-episode psychosis (FEP) and recent onset psychotic disorder (ROP) individuals, with a focus on sex-related differences. Data was collected from PubMed and EMBASE databases. FEP, ROP and high-risk syndromes for psychosis IPD were included if routine basic CSF-diagnostics were reported. Risk of bias of the included studies was evaluated. Random-effects meta-analyses and mixed-effects linear regression models were employed to assess the impact of BCB alterations on symptom severity. Published (6 studies) and unpublished IPD from n = 531 individuals was included in the analyses. CSF was altered in 38.8 % of individuals. No significant differences in symptom severity were found between individuals with and without CSF alterations (SMD = -0.17, 95 %CI -0.55-0.22, p = 0.341). However, males with elevated CSF/serum albumin ratios or any CSF alteration had significantly higher positive symptom scores than those without alterations (SMD = 0.34, 95 %CI 0.05-0.64, p = 0.037 and SMD = 0.29, 95 %CI 0.17-0.41p = 0.005, respectively). Mixed-effects and simple regression models showed no association (p > 0.1) between CSF parameters and symptomatic outcomes. No interaction between sex and CSF parameters was found (p > 0.1). BCB disruption appears highly prevalent in early psychosis and could be involved in positive symptomś severity in males, indicating potential difficult-to-treat states. This work highlights the need for considering BCB breakdownand sex-related differences in SSDs clinical trials and treatment strategies.
Collapse
Affiliation(s)
- Mattia Campana
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Nussbaumstraße 7, D-80336 Munich, Germany.
| | - Vladislav Yakimov
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Nussbaumstraße 7, D-80336 Munich, Germany
| | - Joanna Moussiopoulou
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Nussbaumstraße 7, D-80336 Munich, Germany
| | - Isabel Maurus
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Nussbaumstraße 7, D-80336 Munich, Germany
| | - Lisa Löhrs
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Nussbaumstraße 7, D-80336 Munich, Germany
| | - Florian Raabe
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Nussbaumstraße 7, D-80336 Munich, Germany; Max Planck Institute of Psychiatry, Munich, Germany
| | - Iris Jäger
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Nussbaumstraße 7, D-80336 Munich, Germany
| | - Matin Mortazavi
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, BKH Augsburg, Augsburg, Germany
| | - Michael E Benros
- Copenhagen Research Centre for Biological and Precision Psychiatry. Mental Health Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Rose Jeppesen
- Copenhagen Research Centre for Biological and Precision Psychiatry. Mental Health Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Gerd Meyer Zu Hörste
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Michael Heming
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Eloi Giné-Servén
- Department of Psychiatry, Hospital Universitario de Navarra, Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Javier Labad
- Department of Mental Health, Hospital de Mataró, Consorci Sanitari del Maresme, Mataró, Spain; Translational Neuroscience Research Unit I3PT-INc-UAB, Institut de Innovació i Investigació Parc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, Spain
| | - Ester Boix
- Department of Mental Health, Hospital de Mataró, Consorci Sanitari del Maresme, Mataró, Spain
| | - Belinda Lennox
- Department of Psychiatry, University of Oxford and Oxford Health NHS Foundation Trust, Oxford, UK
| | - Ksenija Yeeles
- Department of Psychiatry, University of Oxford and Oxford Health NHS Foundation Trust, Oxford, UK
| | - Johann Steiner
- Department of Psychiatry, Magdeburg University Hospital, Magdeburg, Germany
| | | | - Henrik Dobrowolny
- Department of Psychiatry, Magdeburg University Hospital, Magdeburg, Germany
| | - Berend Malchow
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Niels Hansen
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Nussbaumstraße 7, D-80336 Munich, Germany; Max Planck Institute of Psychiatry, Munich, Germany; DZPG (German Center for Mental Health), partner site München/Augsburg, Germany
| | - Spyridon Siafis
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University Munich, Munich, Germany
| | - Stefan Leucht
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University Munich, Munich, Germany
| | - Sean Halstead
- Department of Psychiatry, School of Medicine, University of Queensland, Brisbane, Australia
| | - Nicola Warren
- Department of Psychiatry, School of Medicine, University of Queensland, Brisbane, Australia
| | - Dan Siskind
- Department of Psychiatry, School of Medicine, University of Queensland, Brisbane, Australia
| | - Wolfgang Strube
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, BKH Augsburg, Augsburg, Germany
| | - Alkomiet Hasan
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, BKH Augsburg, Augsburg, Germany; DZPG (German Center for Mental Health), partner site München/Augsburg, Germany
| | - Elias Wagner
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Nussbaumstraße 7, D-80336 Munich, Germany; Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, BKH Augsburg, Augsburg, Germany; Evidence-based Psychiatry and Psychotherapy, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| |
Collapse
|
9
|
Abady MM, Jeong JS, Kwon HJ, Assiri AM, Cho J, Saadeldin IM. The reprotoxic adverse side effects of neurogenic and neuroprotective drugs: current use of human organoid modeling as a potential alternative to preclinical models. Front Pharmacol 2024; 15:1412188. [PMID: 38948466 PMCID: PMC11211546 DOI: 10.3389/fphar.2024.1412188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/29/2024] [Indexed: 07/02/2024] Open
Abstract
The management of neurological disorders heavily relies on neurotherapeutic drugs, but notable concerns exist regarding their possible negative effects on reproductive health. Traditional preclinical models often fail to accurately predict reprotoxicity, highlighting the need for more physiologically relevant systems. Organoid models represent a promising approach for concurrently studying neurotoxicity and reprotoxicity, providing insights into the complex interplay between neurotherapeutic drugs and reproductive systems. Herein, we have examined the molecular mechanisms underlying neurotherapeutic drug-induced reprotoxicity and discussed experimental findings from case studies. Additionally, we explore the utility of organoid models in elucidating the reproductive complications of neurodrug exposure. Have discussed the principles of organoid models, highlighting their ability to recapitulate neurodevelopmental processes and simulate drug-induced toxicity in a controlled environment. Challenges and future perspectives in the field have been addressed with a focus on advancing organoid technologies to improve reprotoxicity assessment and enhance drug safety screening. This review underscores the importance of organoid models in unraveling the complex relationship between neurotherapeutic drugs and reproductive health.
Collapse
Affiliation(s)
- Mariam M. Abady
- Organic Metrology Group, Division of Chemical and Material Metrology, Korea Research Institute of Standards and Science, Daejeon, Republic of Korea
- Department of Bio-Analytical Science, University of Science and Technology, Daejeon, Republic of Korea
- Department of Nutrition and Food Science, National Research Centre, Cairo, Egypt
| | - Ji-Seon Jeong
- Organic Metrology Group, Division of Chemical and Material Metrology, Korea Research Institute of Standards and Science, Daejeon, Republic of Korea
- Department of Bio-Analytical Science, University of Science and Technology, Daejeon, Republic of Korea
| | - Ha-Jeong Kwon
- Organic Metrology Group, Division of Chemical and Material Metrology, Korea Research Institute of Standards and Science, Daejeon, Republic of Korea
| | - Abdullah M. Assiri
- Deperament of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Jongki Cho
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Republic of Korea
| | - Islam M. Saadeldin
- Deperament of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
10
|
Elmorsy E, Al-Ghafari A, Al Doghaither H, Hashish S, Salama M, Mudyanselage AW, James L, Carter WG. Differential Effects of Paraquat, Rotenone, and MPTP on Cellular Bioenergetics of Undifferentiated and Differentiated Human Neuroblastoma Cells. Brain Sci 2023; 13:1717. [PMID: 38137165 PMCID: PMC10741680 DOI: 10.3390/brainsci13121717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Paraquat (PQ), rotenone (RO), and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) are neurotoxicants that can damage human health. Exposure to these neurotoxicants has been linked to neurodegeneration, particularly Parkinson's disease. However, their mechanisms of action have not been fully elucidated, nor has the relative vulnerability of neuronal subtypes to their exposures. To address this, the current study investigated the cytotoxic effects of PQ, RO, and MPTP and their relative effects on cellular bioenergetics and oxidative stress on undifferentiated human neuroblastoma (SH-SY5Y) cells and those differentiated to dopaminergic (DA) or cholinergic (CH) phenotypes. The tested neurotoxicants were all cytotoxic to the three cell phenotypes that correlated with both concentration and exposure duration. At half-maximal effective concentrations (EC50s), there were significant reductions in cellular ATP levels and reduced activity of the mitochondrial complexes I and III, with a parallel increase in lactate production. PQ at 10 µM significantly decreased ATP production and mitochondrial complex III activity only in DA cells. RO was the most potent inhibitor of mitochondrial complex 1 and did not inhibit mitochondrial complex III even at concentrations that induced a 50% loss of cell viability. MPTP was the most potent toxicant in undifferentiated cells. All neurotoxicants significantly increased reactive oxygen species, lipid peroxidation, and nuclear expression of Nrf2, with a corresponding inhibition of the antioxidant enzymes catalase and superoxide dismutase. At a 10 µM exposure to PQ or RO, oxidative stress biomarkers were significant in DA cells. Collectively, this study underscores the importance of mitochondrial dysfunction and oxidative stress in PQ, RO, and MPTP-induced cytotoxicity and that neuronal phenotypes display differential vulnerability to these neurotoxicants.
Collapse
Affiliation(s)
- Ekramy Elmorsy
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
- Pathology Department, Faculty of Medicine, Northern Border University, Arar 91431, Saudi Arabia
| | - Ayat Al-Ghafari
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.A.-G.); (H.A.D.)
- Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Huda Al Doghaither
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.A.-G.); (H.A.D.)
| | - Sara Hashish
- Institute of Global Health and Human Ecology, The American University in Cairo (AUC), Cairo 11385, Egypt; (S.H.); (M.S.)
| | - Mohamed Salama
- Institute of Global Health and Human Ecology, The American University in Cairo (AUC), Cairo 11385, Egypt; (S.H.); (M.S.)
| | - Anusha W. Mudyanselage
- Clinical Toxicology Research Group, School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby DE22 3DT, UK; (A.W.M.); (L.J.)
- Faculty of Agricultural Sciences, Sabaragamuwa University of Sri Lanka, Belihuloya 70140, Sri Lanka
| | - Lipta James
- Clinical Toxicology Research Group, School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby DE22 3DT, UK; (A.W.M.); (L.J.)
| | - Wayne G. Carter
- Clinical Toxicology Research Group, School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby DE22 3DT, UK; (A.W.M.); (L.J.)
| |
Collapse
|
11
|
Elmorsy EM, Al-Ghafari A, Al Doghaither H. Role of mitochondrial disruption and oxidative stress in plasticizer phthalate-induced cytotoxicity to human bone osteoblasts. Toxicol Res (Camb) 2023; 12:765-774. [PMID: 37915493 PMCID: PMC10615828 DOI: 10.1093/toxres/tfad065] [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: 03/22/2023] [Revised: 06/13/2023] [Accepted: 07/25/2023] [Indexed: 11/03/2023] Open
Abstract
Phthalates are frequently utilized in a wide range of products such as plasticizers with reported negative effects on bones. The current study evaluated the effect of butyl cyclohexyl phthalate on the human osteoblasts via different assays. MTT and lactate dehydrogenase assays were used to examine the in-vitro cytotoxic effect of butyl cyclohexyl phthalate on human bone osteoblasts in concentrations 0.1, 1, 10, and 100 μM for 12 to 72 h postexposures. Incubation of osteoblasts with butyl cyclohexyl phthalate significantly reduced cell viability based on its concentrations and durations of exposure. In parallel, osteoblast secretion of procollagen type 1, osteocalcin, as well as alkaline phosphatase was significantly decreased by butyl cyclohexyl phthalate in concentrations (1 or 2 μM). Butyl cyclohexyl phthalate decreased ATP synthesis and mitochondrial complexes I and III activities, with increased lactate production, all of which were detrimental to cellular bioenergetics. The cellular redox defense systems were significantly depleted by increased lipid peroxidation, elevated reactive oxygen species, decreased catalase and superoxide dismutase enzymes activities, and decreased intracellular reduced glutathione (GSH). Redox stress was also induced. Interestingly, preincubating osteoblasts with reduced GSH before exposing them to butyl cyclohexyl phthalate significantly lowered the cytotoxicity of the butyl cyclohexyl phthalate, suggesting that antioxidants may play a helpful protective effect.
Collapse
Affiliation(s)
- Ekramy Mahmoud Elmorsy
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
- Pathology Department, Faculty of Medicine, Northern Border University, Arar 1321, Saudi Arabia
| | - Ayat Al-Ghafari
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah 80200, Saudi Arabia
- Cancer Metabolism and Epigenetics Unit, Faculty of Science, King Abdulaziz University, Jeddah 80200, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah 80200, Saudi Arabia
- Cancer and Mutagenesis Research Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah 80200, Saudi Arabia
| | - Huda Al Doghaither
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah 80200, Saudi Arabia
| |
Collapse
|
12
|
Ngamratanapaiboon S, Srikornvit N, Hongthawonsiri P, Pornchokchai K, Wongpitoonmanachai S, Pholkla P, Mo J, Yambangyang P, Ayutthaya WDN. Exploring the mechanisms of clozapine-induced blood-brain barrier dysfunction using untargeted metabolomics and cellular metabolism analysis. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 102:104219. [PMID: 37451530 DOI: 10.1016/j.etap.2023.104219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/24/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Brain microvascular endothelial cells (BMVECs) from the blood- brain barrier form a highly selective membrane that protects the brain from circulating blood and maintains a stable microenvironment for the central nervous system. BMVEC dysfunction has been implicated in a variety of neurological and psychiatric disorders. Clozapine, a widely used antipsychotics, has been demonstrated to alter the permeability of BMVECs, but the underlying mechanisms of this effect are not fully understood. In this study, we investigated the effects of clozapine in BMVECs using untargeted metabolomics analysis. Our results illustrated that treatment with clozapine led to significant changes in the metabolic profile of BMVECs, including alterations in amino acid and energy metabolism. These findings suggest that clozapine affects BMVEC permeability through its effects on cellular metabolism. Our study could inform the development of more targeted and effective treatments for understanding the relationships among clozapine, cellular metabolism, and BMVECs in more detail.
Collapse
Affiliation(s)
- Surachai Ngamratanapaiboon
- Division of Pharmacology, Department of Basic Medical Sciences, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok 10300, Thailand.
| | - Napatarin Srikornvit
- Medical Student in Doctor of Medicine Programme, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok 10300, Thailand
| | - Patipol Hongthawonsiri
- Medical Student in Doctor of Medicine Programme, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok 10300, Thailand
| | - Krittaboon Pornchokchai
- Medical Student in Doctor of Medicine Programme, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok 10300, Thailand
| | - Siriphattarinya Wongpitoonmanachai
- Medical Student in Doctor of Medicine Programme, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok 10300, Thailand
| | - Petchlada Pholkla
- Medical Student in Doctor of Medicine Programme, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok 10300, Thailand
| | - Jiajun Mo
- Medical Student in Doctor of Medicine Programme, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok 10300, Thailand
| | - Pracha Yambangyang
- Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Salaya, Nakhon Pathom 73170, Thailand
| | - Watcharaporn Devakul Na Ayutthaya
- Division of Pharmacology, Department of Basic Medical Sciences, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok 10300, Thailand
| |
Collapse
|
13
|
Tonyali A, Karacetin G, Yesilkaya C, Arik FNT, Kayan Ocakoglu B. Appearance of extrapyramidal symptoms in adolescent psychiatry patients during COVID-19 infection. J Med Virol 2023; 95:e28556. [PMID: 36738231 DOI: 10.1002/jmv.28556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/08/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
Since the start of the pandemic, there has been an increase in the incidence of psychiatric morbidity among those infected with coronavirus disease 2019 (COVID-19) and those indirectly affected by COVID-19. There has been a considerable increase in the number of individuals with such psychiatric conditions as depression, acute stress disorders, anxiety, and posttraumatic stress disorder (PTSD). About one-third of patients with COVID-19 are reported to have developed short and long-term neuropsychiatric conditions such as delirium, agitation, altered consciousness, hypoxic encephalopathy encephalitis, dysexecutive syndrome, cerebrovascular complications (e.g., stroke), hypoxic encephalopathy, convulsions, neuromuscular dysfunction, demyelinating processes, or parkinsonism through several pathophysiological mechanisms. Nevertheless, as the pandemic progressed, data on neuropsychiatric manifestations implied that the pathologic capacity of COVID-19 and its association with the onset and/or exacerbation of psychiatric morbidity indicate that COVID-19 is potentially related to neuropsychiatric involvement. Patients with existing mental disorders under psychotropic treatment exposed to the COVID-19 infection have been represented by an increased risk of worsened psychiatric symptoms and expanded drug side effects. The present study aimed to describe five pediatric patients with various psychiatric illness that experienced COVID-19 infection and had potentially associated neuropsychiatric involvement, such as exacerbation of underlying psychiatric symptoms and extrapyramidal side effects. To the best of our knowledge, the present study is the first to describe adolescents with COVID-19 infection that presented with a series of manifestations in the form of an increase in extrapyramidal symptoms (EPS) during exacerbation of underlying psychiatric disease.
Collapse
Affiliation(s)
- Aysegul Tonyali
- Istanbul Bakırköy Prof. Dr. Mazhar Osman Training and Research Hospital, Istanbul, Turkey
| | | | | | | | | |
Collapse
|
14
|
Rabkin SW, Tang JKK. Clozapine-induced Myocarditis: Pathophysiologic Mechanisms and Implications for Therapeutic Approaches. Curr Mol Pharmacol 2023; 16:60-70. [PMID: 35152873 DOI: 10.2174/1874467215666220211094910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 11/19/2021] [Accepted: 11/29/2021] [Indexed: 11/22/2022]
Abstract
Clozapine, a superior treatment for treatment-resistant schizophrenia can cause potentially life-threatening myocarditis and dilated cardiomyopathy. While the occurrence of this condition is well known, its molecular mechanisms are unclear and may be multifactorial. Putative mechanisms warrant an in-depth review not only from the perspective of toxicity but also for understanding the molecular mechanisms of the adverse cardiac effects of clozapine and the development of novel therapeutic approaches. Clozapine-induced cardiac toxicity encompasses a diverse set of pathways, including (i) immune modulation and proinflammatory processes encompassing an IgEmediated (type I hypersensitivity) response and perhaps a cytokine release syndrome (ii) catecholaminergic activation (iii) induction of free radicals and oxidative stress (iv) activation of cardiomyocyte cell death pathways, including apoptosis, ischemia through impairment in coronary blood flow via changes in endothelial production of NO and vasoconstriction induced by norepinephrine as well as other factors released from cardiac mast cells. (v) In addition, an extensive examination of the effects of clozapine on non-cardiac cellular proteins demonstrates that clozapine can impair enzymes involved in cellular metabolism, such as pyruvate kinase, mitochondrial malate dehydrogenase, and other proteins, including α-enolase, triosephosphate isomerase and cofilin, which might explain clozapine-induced reductions in myocardial energy generation for cell viability as well as contractile function. Pharmacologic antagonism of these cellular protein effects may lead to the development of strategies to antagonize the cardiac damage induced by clozapine.
Collapse
Affiliation(s)
- Simon W Rabkin
- Division of Cardiology, University of British Columbia, Vancouver, B.C., Canada
| | - Jacky K K Tang
- Division of Cardiology, University of British Columbia, Vancouver, B.C., Canada
| |
Collapse
|
15
|
Omar UM, Elmorsy EM, Al-Ghafari AB. Mitochondrial disruption in isolated human monocytes: an underlying mechanism for cadmium-induced immunotoxicity. J Immunotoxicol 2022; 19:81-92. [PMID: 36067115 DOI: 10.1080/1547691x.2022.2113840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cadmium (Cd) is an immunotoxic metal frequently found in the environment. The in vitro study undertaken here evaluated the immunotoxic effects of Cd in isolated human peripheral blood monocytes (hPBM). The results of the studies of exposures to varying doses of Cd (0, 0.1, 1, 10, and 100 µM, as cadmium dichloride [CdCl2]) for 3, 6, 12, 24, 48, and 72 hr showed the test agent was cytotoxic to the cells in time- and concentration-related manners. Thereafter, using only those doses found to not cause extreme cell lethality a 48-hr period, the impact of 0.1 or 1 µM CdCl2 on the cells was evaluated. Functionally, CdCl2 treatment led to time- and concentration-related decreases in hPBM phagocytic activities as well as in the ability of the cells to form/release cytokines (including tumor necrosis factor [TNF]-α and interleukin [IL]-6 and -8). The CdCl2 also led to significantly decreased ATP production (in part, via inhibition of mitochondrial complexes I and III) as well as in mitochondrial membrane potentials (MMP) and oxygen consumption rates (OCR; associated with parallel increases in cell lactate production) in the cells. In addition, CdCl2 treatment resulted in significant increases in mitochondrial membrane fluidity (MMF) and cell unsaturated fatty acid content. Based on the results here, one might conclude that some of the effects that arose during the CdCl2-induced dysfunction of the isolated hPBM (i.e. changes phagocytic activity, cytokine formation/secretion) could have evolved secondary to CdCl2-induced disruptions of hPBM cell bioenergetics - an effect that itself was a culmination of an overall toxicity from CdCl2 upon the mitochondria within these cells.
Collapse
Affiliation(s)
- Ulfat M Omar
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ekramy M Elmorsy
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.,Pathology Department, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia
| | - Ayat B Al-Ghafari
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.,Cancer and Mutagenesis Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Research Center, Dar Al-Hekma University, Jeddah, Saudi Arabia
| |
Collapse
|
16
|
Yazla E, Kayadibi H, Cetin I, Aydinoglu U, Karadere ME. Evaluation of Changes in Peripheric Biomarkers Related to Blood Brain Barrier Damage in Patients with Schizophrenia and Their Correlation with Symptoms. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2022; 20:504-513. [PMID: 35879035 PMCID: PMC9329119 DOI: 10.9758/cpn.2022.20.3.504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/06/2021] [Accepted: 06/18/2021] [Indexed: 11/18/2022]
Abstract
Objective The aim of the study was to evaluate the levels of peripheric biomarkers that have been associated with blood brain barrier (BBB) damage in healthy controls and two groups of patients with schizophrenia, those who received typical-atypical antipsychotics and those who received only atypical antipsychotics. Additionally, we sought relationships between these biomarkers and schizophrenia symptoms. Methods This study was conducted with the inclusion of 41 healthy volunteers and 75 patients with schizophrenia. The biomarkers measured to evaluate BBB injury were as follows spectrin breakdown product 145 (SBDP145), spectrin breakdown product 150 (SBDP150), ubiquitin carboxy terminal hydrolase L1 (UCHL1), ubiquitin ligase cullin-3 (cullin), occludin and claudin, which were measured via ELISA. Symptoms of patients with schizophrenia were evaluated with the Scale for the Assessment of Positive Symptoms (SAPS), Scale for the Assessment of Negative Symptoms, the Clinical Global Impression Scale (CGI), and the general assessment of functionality (GAF). Results Compared to controls, SBDP145 (p = 0.022) and cullin (p = 0.046) levels were significantly higher in patients with schizophrenia receiving atypical antipsychotic treatment. SBDP150 levels were lower in the combination treatment group compared to the control group (p = 0.022). Claudin (p = 0.804), occludin (p = 0.058) and UCHL1 (p = 0.715) levels were similar among groups. In recipients of combination treatment, SBDP145 levels were found to be positively correlated with SAPS-total (r = 0.440, p = 0.036) and SAPS-delusions (r = 0.494, p = 0.017) scores. Conclusion The relationships demonstrated in this study indicate that more comprehensive research is needed to understand whether BBB defects contribute to clinical characteristics in patients with schizophrenia.
Collapse
Affiliation(s)
- Ece Yazla
- Department of Psychiatry, Hitit University Faculty of Medicine, Corum, Turkey
| | - Huseyin Kayadibi
- Department of Biochemistry, Eskisehir Osmangazi University Faculty of Medicine, Eskisehir, Turkey
| | - Ihsan Cetin
- Department of Biochemistry, Hitit University Faculty of Medicine, Corum, Turkey
| | - Unsal Aydinoglu
- Department of Psychiatry, Hitit University Faculty of Medicine, Corum, Turkey
| | - Mehmet Emrah Karadere
- Department of Psychiatry, Istanbul Medeniyet University Faculty of Medicine, Istanbul, Turkey
| |
Collapse
|
17
|
de Bartolomeis A, Vellucci L, Barone A, Manchia M, De Luca V, Iasevoli F, Correll CU. Clozapine's multiple cellular mechanisms: What do we know after more than fifty years? A systematic review and critical assessment of translational mechanisms relevant for innovative strategies in treatment-resistant schizophrenia. Pharmacol Ther 2022; 236:108236. [PMID: 35764175 DOI: 10.1016/j.pharmthera.2022.108236] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 12/21/2022]
Abstract
Almost fifty years after its first introduction into clinical care, clozapine remains the only evidence-based pharmacological option for treatment-resistant schizophrenia (TRS), which affects approximately 30% of patients with schizophrenia. Despite the long-time experience with clozapine, the specific mechanism of action (MOA) responsible for its superior efficacy among antipsychotics is still elusive, both at the receptor and intracellular signaling level. This systematic review is aimed at critically assessing the role and specific relevance of clozapine's multimodal actions, dissecting those mechanisms that under a translational perspective could shed light on molecular targets worth to be considered for further innovative antipsychotic development. In vivo and in vitro preclinical findings, supported by innovative techniques and methods, together with pharmacogenomic and in vivo functional studies, point to multiple and possibly overlapping MOAs. To better explore this crucial issue, the specific affinity for 5-HT2R, D1R, α2c, and muscarinic receptors, the relatively low occupancy at dopamine D2R, the interaction with receptor dimers, as well as the potential confounder effects resulting in biased ligand action, and lastly, the role of the moiety responsible for lipophilic and alkaline features of clozapine are highlighted. Finally, the role of transcription and protein changes at the synaptic level, and the possibility that clozapine can directly impact synaptic architecture are addressed. Although clozapine's exact MOAs that contribute to its unique efficacy and some of its severe adverse effects have not been fully understood, relevant information can be gleaned from recent mechanistic understandings that may help design much needed additional therapeutic strategies for TRS.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment Resistant Psychosis, Department of Neuroscience, Reproductive Science and Dentistry, University Medical School of Naples "Federico II", Naples, Italy.
| | - Licia Vellucci
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment Resistant Psychosis, Department of Neuroscience, Reproductive Science and Dentistry, University Medical School of Naples "Federico II", Naples, Italy
| | - Annarita Barone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment Resistant Psychosis, Department of Neuroscience, Reproductive Science and Dentistry, University Medical School of Naples "Federico II", Naples, Italy
| | - Mirko Manchia
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy; Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | - Felice Iasevoli
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment Resistant Psychosis, Department of Neuroscience, Reproductive Science and Dentistry, University Medical School of Naples "Federico II", Naples, Italy
| | - Christoph U Correll
- The Zucker Hillside Hospital, Department of Psychiatry, Northwell Health, Glen Oaks, NY, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Department of Psychiatry and Molecular Medicine, Hempstead, NY, USA; Charité Universitätsmedizin Berlin, Department of Child and Adolescent Psychiatry, Berlin, Germany
| |
Collapse
|
18
|
Cheng Y, Wang T, Zhang T, Yi S, Zhao S, Li N, Yang Y, Zhang F, Xu L, Shan B, Xu X, Xu J. Increased blood-brain barrier permeability of the thalamus and the correlation with symptom severity and brain volume alterations in schizophrenia patients. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2022; 7:1025-1034. [PMID: 35738480 DOI: 10.1016/j.bpsc.2022.06.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND Cumulative evidence of microvascular dysfunction has suggested the blood-brain barrier (BBB) disruption in schizophrenia, while the direct in vivo evidence from patients is inadequate. In this study, using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) methods, we tried to test the hypothesis that there was increased BBB permeability in schizophrenia patients, and correlated with the clinical characters, and brain volumetric alterations. METHODS Structural MRI and DCE-MRI data from 29 schizophrenia patients and 18 age- and sex- matched controls (HC) were obtained. We calculated the volume transfer constant (Ktrans) value and compared the difference between two groups. The regions with the abnormal Ktrans value were extracted as ROIs (thalamus), and the correlation with the clinical characters and grey matter volume were analysed. RESULTS The results revealed that, compared with the HC, the volume transfer constant (Ktrans) value of the bilateral thalamus in the schizophrenia group was increased (p < 0.001). There were significant positive correlations between thalamic mean Ktrans value with disease duration (p < 0.05) and symptom severity (p < 0.001). Analysis of the thalamic subregions revealed that the BBB disruption was significant in pulvinar, especially the medial pulvinar nucleus (PuM) and lateral pulvinar nucleus (PuL) (p < 0.001). The Ktrans value of the whole brain, thalamus, and thalamic subregions was negatively correlated with their volume separately. CONCLUSION These results provided the first in vivo evidence of BBB disruption of thalamus in schizophrenia patients, and the BBB dysfunction might contribute to the pathological brain structural alterations in schizophrenia.
Collapse
Affiliation(s)
- Yuqi Cheng
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming, China, 650032; Yunnan Clinical Research Centre for Mental Health, Kunming, China, 650032.
| | - Ting Wang
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming, China, 650032
| | - Tianhao Zhang
- Laboratory of Nuclear Analysis Techniques, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049
| | - Shu Yi
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming, China, 650032
| | - Shilun Zhao
- Laboratory of Nuclear Analysis Techniques, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049
| | - Na Li
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming, China, 650032
| | - Yifan Yang
- Department of Rheumatology, First Affiliated Hospital of Kunming Medical University, Kunming, China, 650032
| | - Fengrui Zhang
- Department of Medical Imaging, First Affiliated Hospital of Kunming Medical University, Kunming, China, 650032
| | - Lin Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, China, 650223
| | - Baoci Shan
- Laboratory of Nuclear Analysis Techniques, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049
| | - Xiufeng Xu
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming, China, 650032
| | - Jian Xu
- Department of Rheumatology, First Affiliated Hospital of Kunming Medical University, Kunming, China, 650032.
| |
Collapse
|
19
|
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
|
20
|
Zhao NO, Topolski N, Tusconi M, Salarda EM, Busby CW, Lima CN, Pillai A, Quevedo J, Barichello T, Fries GR. Blood-brain barrier dysfunction in bipolar disorder: Molecular mechanisms and clinical implications. Brain Behav Immun Health 2022; 21:100441. [PMID: 35308081 PMCID: PMC8924633 DOI: 10.1016/j.bbih.2022.100441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 12/02/2022] Open
Abstract
Bipolar disorder (BD) is a severe psychiatric disorder affecting approximately 1-3% of the population and characterized by a chronic and recurrent course of debilitating symptoms. An increasing focus has been directed to discover and explain the function of Blood-Brain Barrier (BBB) integrity and its association with a number of psychiatric disorders; however, there has been limited research in the role of BBB integrity in BD. Multiple pathways may play crucial roles in modulating BBB integrity in BD, such as inflammation, insulin resistance, and alterations of neuronal plasticity. In turn, BBB impairment is hypothesized to have a significant clinical impact in BD patients. Based on the high prevalence of medical and psychiatric comorbidities in BD and a growing body of evidence linking inflammatory and neuroinflammatory mechanisms to the disorder, recent studies have suggested that BBB dysfunction may play a key role in BD's pathophysiology. In this comprehensive narrative review, we aim to discuss studies investigating biological markers of BBB in patients with BD, mechanisms that modulate BBB integrity, their clinical implications on patients, and key targets for future development of novel therapies.
Collapse
Affiliation(s)
- Ning O. Zhao
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, 1941 East Rd, 77054, Houston, TX, USA
| | - Natasha Topolski
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, 1941 East Rd, 77054, Houston, TX, USA
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX. 6767 Bertner Ave, 77030, Houston, TX, USA
| | - Massimo Tusconi
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Italy
| | - Erika M. Salarda
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, 1941 East Rd, 77054, Houston, TX, USA
| | - Christopher W. Busby
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, 1941 East Rd, 77054, Houston, TX, USA
| | - Camila N.N.C. Lima
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, 1941 East Rd, 77054, Houston, TX, USA
| | - Anilkumar Pillai
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, 1941 East Rd, 77054, Houston, TX, USA
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX. 6767 Bertner Ave, 77030, Houston, TX, USA
- Pathophysiology of Neuropsychiatric Disorders Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Joao Quevedo
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, 1941 East Rd, 77054, Houston, TX, USA
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX. 6767 Bertner Ave, 77030, Houston, TX, USA
| | - Tatiana Barichello
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, 1941 East Rd, 77054, Houston, TX, USA
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX. 6767 Bertner Ave, 77030, Houston, TX, USA
| | - Gabriel R. Fries
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, 1941 East Rd, 77054, Houston, TX, USA
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX. 6767 Bertner Ave, 77030, Houston, TX, USA
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA. 7000 Fannin, 77030, Houston, TX, USA
| |
Collapse
|
21
|
Ngamratanapaiboon S, Yambangyang P. Quantification of antipsychotic biotransformation in brain microvascular endothelial cells by using untargeted metabolomics. Drug Discov Ther 2022; 15:317-324. [PMID: 35034925 DOI: 10.5582/ddt.2021.01101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Most studies of antipsychotic-therapies have highlighted the discrepancy between plasma and brain pharmacokinetics of antipsychotics, but how the drug changes through the blood brain barrier (BBB) has not been investigated. Cell-based metabolomics using liquid chromatography-mass spectrometry (LC-MS) combined with multivariate data analysis were applied for screening of antipsychotic metabolites in the BBB. We applied this approach to analyze the antipsychotic biotransformation in brain microvascular endothelia cells (BMVECs), the main component of the BBB. From this study, five, four, three, and one metabolite of chlorpromazine, clozapine, haloperidol and risperidone, respectively, were locally metabolized on the BMVECs. These results confirm that there is a drug biotransformation process within the BBB and show that drug metabolite screening employed cell-based metabolomics using LC-MS, combined with multivariate analysis in the study of BMVECs exposed to antipsychotics can provide a way to screen drug metabolites in the BBB.
Collapse
Affiliation(s)
- Surachai Ngamratanapaiboon
- Division of Pharmacology, Department of Basic Medical Science, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Bangkok, Thailand
| | - Pracha Yambangyang
- Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakhon Pathom, Thailand
| |
Collapse
|
22
|
Matteoni S, Matarrese P, Ascione B, Ricci-Vitiani L, Pallini R, Villani V, Pace A, Paggi MG, Abbruzzese C. Chlorpromazine induces cytotoxic autophagy in glioblastoma cells via endoplasmic reticulum stress and unfolded protein response. J Exp Clin Cancer Res 2021; 40:347. [PMID: 34740374 PMCID: PMC8569984 DOI: 10.1186/s13046-021-02144-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 10/18/2021] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM; grade IV glioma) is characterized by a very short overall survival time and extremely low 5-year survival rates. We intend to promote experimental and clinical research on rationale and scientifically driven drug repurposing. This may represent a safe and often inexpensive way to propose novel pharmacological approaches to GBM. Our precedent work describes the role of chlorpromazine (CPZ) in hindering malignant features of GBM. Here, we investigate in greater detail the molecular mechanisms at the basis of the effect of CPZ on GBM cells. METHODS We employed proteomics platforms, i.e., activity-based protein profiling plus mass spectrometry, to identify potential cellular targets of the drug. Then, by means of established molecular and cellular biology techniques, we assessed the effects of this drug on GBM cell metabolic and survival pathways. RESULTS The experimental output indicated as putative targets of CPZ several of factors implicated in endoplasmic reticulum (ER) stress, with consequent unfolded protein response (UPR). Such a perturbation culminated in a noticeable reactive oxygen species generation and intense autophagic response that resulted in cytotoxic and abortive effects for six GBM cell lines, three of which growing as neurospheres, while it appeared cytoprotective for the RPE-1 human non-cancer neuro-ectodermal cell line. CONCLUSIONS This discrepancy could be central in explaining the lethal effects of the drug on GBM cells and the relatively scarce cytotoxicity toward normal tissues attributed to this compound. The data presented here offer support to the multicenter phase II clinical trial we have undertaken, which consists of the addition of CPZ to first-line treatment of GBM patients carrying a hypo- or un-methylated MGMT gene, i.e. those characterized by intrinsic resistance to temozolomide.
Collapse
Affiliation(s)
- Silvia Matteoni
- Cellular Networks and Molecular Therapeutic Targets, Proteomics Unit, IRCCS - Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Paola Matarrese
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Barbara Ascione
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Lucia Ricci-Vitiani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Roberto Pallini
- Institute of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University School of Medicine, 00168, Rome, Italy
| | - Veronica Villani
- Neuro-Oncology, IRCCS - Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Andrea Pace
- Neuro-Oncology, IRCCS - Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Marco G Paggi
- Cellular Networks and Molecular Therapeutic Targets, Proteomics Unit, IRCCS - Regina Elena National Cancer Institute, 00144, Rome, Italy.
| | - Claudia Abbruzzese
- Cellular Networks and Molecular Therapeutic Targets, Proteomics Unit, IRCCS - Regina Elena National Cancer Institute, 00144, Rome, Italy.
| |
Collapse
|
23
|
Alenazi SA, Elmorsy E, Al-Ghafari A, El-Husseini A. Effect of amphotericin B-deoxycholate (Fungizone) on the mitochondria of Wistar rats' renal proximal tubules cells. J Appl Toxicol 2021; 41:1620-1633. [PMID: 33740284 DOI: 10.1002/jat.4151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/01/2021] [Accepted: 02/05/2021] [Indexed: 01/02/2023]
Abstract
Amphotericin B-deoxycholate (Fungizone [FZ]) is a widely used potent antimycotic drug in spite of its nephrotoxic effect via different mechanisms. The effect of FZ on renal cell bioenergetics is not clear. The current study evaluated the effect of FZ on the bioenergetics of albino rats' isolated renal proximal tubule cells (PTCs). The cytotoxic effect of FZ on the isolated renal cells was assessed by MTT and lactate dehydrogenase (LDH) assays. The effect of FZ on the PTCs uptake (OAT1 and OCT2) and efflux (P-gp and MRP2) transporters was evaluated. Then, the effect of FZ on mitochondria was assessed by studying complexes I-IV activities, lactate assay, oxygen consumption rates (OCR), and western blotting for all mitochondrial complexes. Moreover, the effect of FZ on mitochondrial membrane fluidity (MMF) and fatty acids composition was evaluated. Additionally, the protective effect of coenzyme q10 was studied. Outcomes showed that FZ was cytotoxic to the PTCs in a concentration and time-dependent patterns. FZ significantly inhibited the studied uptake and efflux tubular transporters with inhibition of the mitochondrial complexes activities and parallel increase in lactate production and decrease in OCRs. Finally, FZ significantly reduced the expression of all mitochondrial complexes in addition to significant increase in MMF and MMFA concentration. Coenzyme Q10 was found to significantly decrease FZ-induced cytotoxicity and transporters impairment in the PTC. FZ significantly inhibits bioenergetics of PTC, which may stimulate the cascade of cell death and clinical nephrotoxicity.
Collapse
Affiliation(s)
- Shehab Ahmed Alenazi
- Department of Pediatrics, Faculty of Medicine, Northern Border University, Arar, Kingdom of Saudi Arabia
| | - Ekramy Elmorsy
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.,Department of Pathology, Faculty of Medicine, Northern Border University, Arar, Kingdom of Saudi Arabia
| | - Ayat Al-Ghafari
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia.,Cancer and Mutagenesis Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Amr El-Husseini
- Division of Nephrology, Bone and Mineral Metabolism, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
24
|
Piantino M, Figarol A, Matsusaki M. Three-Dimensional in vitro Models of Healthy and Tumor Brain Microvasculature for Drug and Toxicity Screening. FRONTIERS IN TOXICOLOGY 2021; 3:656254. [PMID: 35295158 PMCID: PMC8915870 DOI: 10.3389/ftox.2021.656254] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/31/2021] [Indexed: 12/16/2022] Open
Abstract
Tissue vascularization is essential for its oxygenation and the homogenous diffusion of nutrients. Cutting-edge studies are focusing on the vascularization of three-dimensional (3D) in vitro models of human tissues. The reproduction of the brain vasculature is particularly challenging as numerous cell types are involved. Moreover, the blood-brain barrier, which acts as a selective filter between the vascular system and the brain, is a complex structure to replicate. Nevertheless, tremendous advances have been made in recent years, and several works have proposed promising 3D in vitro models of the brain microvasculature. They incorporate cell co-cultures organized in 3D scaffolds, often consisting of components of the native extracellular matrix (ECM), to obtain a micro-environment similar to the in vivo physiological state. These models are particularly useful for studying adverse effects on the healthy brain vasculature. They provide insights into the molecular and cellular events involved in the pathological evolutions of this vasculature, such as those supporting the appearance of brain cancers. Glioblastoma multiform (GBM) is the most common form of brain cancer and one of the most vascularized solid tumors. It is characterized by a high aggressiveness and therapy resistance. Current conventional therapies are unable to prevent the high risk of recurrence of the disease. Most of the new drug candidates fail to pass clinical trials, despite the promising results shown in vitro. The conventional in vitro models are unable to efficiently reproduce the specific features of GBM tumors. Recent studies have indeed suggested a high heterogeneity of the tumor brain vasculature, with the coexistence of intact and leaky regions resulting from the constant remodeling of the ECM by glioma cells. In this review paper, after summarizing the advances in 3D in vitro brain vasculature models, we focus on the latest achievements in vascularized GBM modeling, and the potential applications for both healthy and pathological models as platforms for drug screening and toxicological assays. Particular attention will be paid to discuss the relevance of these models in terms of cell-cell, cell-ECM interactions, vascularization and permeability properties, which are crucial parameters for improving in vitro testing accuracy.
Collapse
Affiliation(s)
- Marie Piantino
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Agathe Figarol
- Institut Jean Lamour, UMR 7198 CNRS, Université de Lorraine, Nancy, France
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan
- *Correspondence: Michiya Matsusaki
| |
Collapse
|
25
|
Al Doghaither H, Elmorsy E, Al-Ghafari A, Ghulam J. Roles of oxidative stress, apoptosis, and inflammation in metal-induced dysfunction of beta pancreatic cells isolated from CD1 mice. Saudi J Biol Sci 2021; 28:651-663. [PMID: 33424352 PMCID: PMC7785459 DOI: 10.1016/j.sjbs.2020.10.056] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/20/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023] Open
Abstract
The diabetogenic effects of metals including lead (Pb), mercury (Hg), cadmium (Cd), and molybdenum (Mo) have been reported with poorly identified underlying mechanisms. The current study assessed the effect of metals on the roles of oxidative stress, apoptosis, and inflammation in beta pancreatic cells isolated from CD-1 mice, via different biochemical assays. Data showed that the tested metals were cytotoxic to the isolated cells with impaired glucose stimulated insulin secretion (GSIS). This was associated with increased reactive oxygen species (ROS) production, lipid peroxidation, antioxidant enzymes activities, active proapoptotic caspase-3 (cas-3), inflammatory cytokines interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α) levels in the intoxicated cells. Furthermore, antioxidant-reduced glutathione (GSH-R), cas-3 inhibitor z-VAD-FMK, IL-6 inhibitor bazedoxifene (BZ), and TNF-α inhibitor etanercept (ET) were found to significantly decrease metal-induced cytotoxicity with improved GSIS in metals' intoxicated cells. In conclusion, oxidative stress, apoptosis, and inflammation can play roles in metals-induced diabetogenic effect.
Collapse
Affiliation(s)
- Huda Al Doghaither
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ekramy Elmorsy
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Pathology Department, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia
| | - Ayat Al-Ghafari
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Cancer Metabolism and Epigenetics Unit, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Cancer and Mutagenesis Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jihan Ghulam
- General Education Department, Dar Al-Hekma University, Jeddah Saudi Arabia
| |
Collapse
|
26
|
Elmorsy E, Alelwani W, Kattan S, Babteen N, Alnajeebi A, Ghulam J, Mosad S. Antipsychotics inhibit the mitochondrial bioenergetics of pancreatic beta cells isolated from CD1 mice. Basic Clin Pharmacol Toxicol 2020; 128:154-168. [PMID: 32860481 DOI: 10.1111/bcpt.13484] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/21/2020] [Accepted: 08/23/2020] [Indexed: 01/14/2023]
Abstract
Antipsychotics (APs) are widely used medications with reported diabetogenic side effects. This study investigated the effect of commonly used APs, namely chlorpromazine (CPZ), haloperidol (HAL) and clozapine, on the bioenergetics of male CD1 mice isolated pancreatic beta cells as an underlying mechanism of their diabetogenic effects. The effect of APs on Alamar blue reduction, adenosine triphosphate (ATP) production and glucose-stimulated insulin secretion (GSIS) of isolated beta cells was evaluated. Then, the effects of APs on the activities of mitochondrial complexes and their common coding genes expression, oxygen consumption rate (OCR), mitochondrial membrane potential (MMP) and lactate production were investigated. The effects of APs on the mitochondrial membrane fluidity (MMF) and mitochondrial membrane fatty acid composition were also examined. Results showed that the tested APs significantly decreased cellular ATP production and GSIS of the beta cells. The APs significantly inhibited the activities of mitochondrial complexes and their coding gene expression, MMP and OCR of the treated cells, with a parallel increase in lactate production to different extents with the different APs. CPZ and HAL showed increased MMF and mitochondrial membrane polyunsaturated fatty acid content. In conclusion, the tested APs-induced mitochondrial disruption can play a role in their diabetogenic side effect.
Collapse
Affiliation(s)
- Ekramy Elmorsy
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.,Pathology Department, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia
| | - Walla Alelwani
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Shahad Kattan
- Medical Laboratory Department, College of Applied Medical Sciences, Taibah University, Yanbu, Saudi Arabia
| | - Nouf Babteen
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Afnan Alnajeebi
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Jihan Ghulam
- General Education Department, Dar Al-Hekma University, Jeddah, Saudi Arabia
| | - Soad Mosad
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.,Pathology Department, Faculty of Medicine, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
27
|
Alelwani W, Elmorsy E, Kattan SW, Babteen NA, Alnajeebi AM, Al-Ghafari A, Carter WG. Carbamazepine induces a bioenergetics disruption to microvascular endothelial cells from the blood-brain barrier. Toxicol Lett 2020; 333:184-191. [PMID: 32805338 DOI: 10.1016/j.toxlet.2020.08.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/15/2020] [Accepted: 08/11/2020] [Indexed: 10/23/2022]
Abstract
Carbamazepine (CBZ) is a widely employed anti-seizure medication that crosses the blood-brain barrier (BBB) to exert its anti-convulsant action. The effects of CBZ on components of the BBB have yet to be completely delineated. Hence the current study evaluated the effects of CBZ upon mitochondrial functionality of BBB-derived microvascular endothelial cells isolated from Albino rats. The influence of CBZ on cell viability and barrier functions were evaluated by 3-(4,5 dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT), lactate dehydrogenase, and electrophysiological assays over a drug concentration range of 0.1-1000 μM. Bioenergetics effects were measured via ATP production, mitochondrial complexes I and III activities, lactate production, and oxygen consumption rates (OCRs), and mitochondrial membrane potential, fluidity and lipid content. CBZ was cytotoxic to microvascular endothelial cells in a concentration and duration dependent manner. CBZ significantly diminished the endothelial cell's barrier functions, and impacted upon cellular bioenergetics: reducing mitochondrial complex activities with a parallel decrease in OCRs and increased anaerobic lactate production. CBZ significantly decreased mitochondrial membrane potential and induced an increase of membrane fluidity and decrease in levels of mitochondrial saturated and unsaturated fatty acids. In summary, CBZ disrupted functional activity of BBB endothelial cells via damage and modification of mitochondria functionality at therapeutically relevant concentrations.
Collapse
Affiliation(s)
- Walla Alelwani
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah 80203, Saudi Arabia
| | - Ekramy Elmorsy
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura, Egypt; School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby, UK
| | - Shahad W Kattan
- Medical Laboratory Department, College of Applied Medical Sciences, Taibah University, Yanbu, Saudi Arabia
| | - Nouf Abubakr Babteen
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah 80203, Saudi Arabia
| | - Afnan M Alnajeebi
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah 80203, Saudi Arabia
| | - Ayat Al-Ghafari
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Wayne G Carter
- School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby, UK.
| |
Collapse
|
28
|
Bouvier ML, Fehsel K, Schmitt A, Meisenzahl-Lechner E, Gaebel W, von Wilmsdorff M. Sex-dependent alterations of dopamine receptor and glucose transporter density in rat hypothalamus under long-term clozapine and haloperidol medication. Brain Behav 2020; 10:e01694. [PMID: 32525610 PMCID: PMC7428470 DOI: 10.1002/brb3.1694] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/04/2020] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE Sex-dependent disturbances of peripheral glucose metabolism are known complications of antipsychotic drug treatment. The influence of long-term clozapine and haloperidol medication on hypothalamus, maintaining aspects of internal body homeostasis, has not yet been completely clarified. METHODS After puberty, male and female Sprague Dawley rats were fed orally with ground pellets containing haloperidol (1 mg/kgBW/day) or clozapine (20 mg/kgBW/day) for 12 weeks. The hypothalamic protein expression of dopamine receptors D2R and D4R, melanocortin receptor MC4R, and glucose transporters Glut1 and Glut3 was examined. Glucose, glycogen, lactate, and pyruvate levels were determined, also malondialdehyde equivalents as markers of oxidative stress. RESULTS D2R expression was increased in the male haloperidol and clozapine group but decreased in females medicated with clozapine. D4R expression was upregulated under clozapine medication. While females showed increased Glut1, Glut3 was elevated in both male and female clozapine-medicated animals. We found no changes of hypothalamic malondialdehyde, glycogen, and MC4R. Hypothalamic lactate was elevated in the female clozapine group. CONCLUSION Clozapine sex-dependently affects the expression of D2R, Glut1, and Glut3. The upregulation of the glucose transporters indicates glucose deprivation in the endothelial cells and consequently in astrocytes and neurons. Increased hypothalamic lactate in females under clozapine points to enhanced glycolysis with a higher glucose demand to produce the required energy. Haloperidol did not change the expression of the glucose transporters and upregulated D2R only in males.
Collapse
Affiliation(s)
- Marie-Luise Bouvier
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Karin Fehsel
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Andrea Schmitt
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilians University Munich, München, Germany.,Laboratory of Neuroscience (LIM27), Institute of Psychiatry, University of Sao Paulo, São Paulo, Brazil
| | - Eva Meisenzahl-Lechner
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Wolfgang Gaebel
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Martina von Wilmsdorff
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
29
|
Silverstein SM, Fradkin SI, Demmin DL. Schizophrenia and the retina: Towards a 2020 perspective. Schizophr Res 2020; 219:84-94. [PMID: 31708400 PMCID: PMC7202990 DOI: 10.1016/j.schres.2019.09.016] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 09/28/2019] [Accepted: 09/30/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Differences between people with schizophrenia and psychiatrically healthy controls have been consistently demonstrated on measures of retinal function such as electroretinography (ERG), and measures of retinal structure such as optical coherence tomography (OCT). Since our 2015 review of this literature, multiple new studies have been published using these techniques. At the same time, the accumulation of data has highlighted the "fault lines" in these fields, suggesting methodological considerations that need greater attention in future studies. METHODS We reviewed studies of ERG and OCT in schizophrenia, as well as data from studies whose findings are relevant to interpreting these papers, such as those on effects of the following on ERG and OCT data: comorbid medical conditions that are over-represented in schizophrenia, smoking, antipsychotic medication, substance abuse, sex and gender, obesity, attention, motivation, and influences of brain activity on retinal function. RESULTS Recent ERG and OCT studies continue to support the hypothesis of retinal structural and functional abnormalities in schizophrenia, and suggest that these are relevant to understanding broader aspects of pathophysiology, neurodevelopment, and neurodegeneration in this disorder. However, there are differences in findings which suggest that the effects of multiple variables on ERG and OCT data need further clarification. CONCLUSIONS The retina, as the only component of the CNS that can be imaged directly in live humans, has potential to clarify important aspects of schizophrenia. With greater attention to specific methodological issues, the true potential of ERG and OCT as biomarkers for important clinical phenomena in schizophrenia should become apparent.
Collapse
Affiliation(s)
- Steven M Silverstein
- Rutgers University Behavioral Health Care, United States; Rutgers University, Robert Wood Johnson Medical School, Departments of Psychiatry and Ophthalmology, United States.
| | | | - Docia L Demmin
- Rutgers University, Department of Psychology, United States.
| |
Collapse
|
30
|
Al-Ghafari A, Elmorsy E, Fikry E, Alrowaili M, Carter WG. The heavy metals lead and cadmium are cytotoxic to human bone osteoblasts via induction of redox stress. PLoS One 2019; 14:e0225341. [PMID: 31756223 PMCID: PMC6874340 DOI: 10.1371/journal.pone.0225341] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/01/2019] [Indexed: 01/09/2023] Open
Abstract
The heavy metals (HMs) lead and cadmium are persistent environmental pollutants capable of inducing ill-health in exposed individuals. One of the primary sites of accumulation and potential damage from HMs is bone, and we therefore examined the acute effects of lead and cadmium on human bone osteoblasts in vitro over a concentration range of 0.1 μM to 1mM, and for 3, 6, 12, 24, and 48 hour exposures. Incubation of osteoblasts with either lead or cadmium reduced cell viability in a concentrations and exposure durations dependent manner, as measured using MTT and LDH assays. Cytotoxicity was significant from 0.1 μM concentrations after 48 hour exposures. Both HMs damaged cellular bioenergetics with reductions of ATP production, mitochondrial complex activities, and aerobic respiration. There was a concomitant elevation of reactive oxygen species, with induction of redox stress measured as increased lipid peroxidation, and depleted cellular redox defense systems via reduced superoxide dismutase and catalase activity and cellular glutathione levels. Both HMs induced nuclear activation of Nrf2, presumably to increase transcription of antioxidant responsive genes to combat oxidative stress. Incubation of osteoblasts with HMs also compromised the secretion of procollagen type 1, osteocalcin, and alkaline phosphatase. Pre-incubation of osteoblasts with reduced glutathione prior to challenge with HMs lessened the cytotoxicity of the HMs, indicative that antioxidants may be a beneficial treatment adjunct in cases of acute lead or cadmium poisoning.
Collapse
Affiliation(s)
- Ayat Al-Ghafari
- Biochemistry Department, Faculty of Science, King AbdulAziz University, Jeddah, Saudi Arabia
| | - Ekramy Elmorsy
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura City, Egypt.,Department of Pathology, Faculty of Medicine, Northern Border University, Arar; Saudi Arabia.,School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby, United Kingdom
| | - Emad Fikry
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura City, Egypt
| | - Majed Alrowaili
- Department of Surgery, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia
| | - Wayne G Carter
- School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby, United Kingdom
| |
Collapse
|
31
|
Long-Lasting Symptomatic Cerebral Hyperperfusion Syndrome following Superficial Temporal Artery-Middle Cerebral Artery Bypass in a Patient with Stenosis of Middle Cerebral Artery. Case Rep Neurol Med 2018; 2018:4717256. [PMID: 30345131 PMCID: PMC6174737 DOI: 10.1155/2018/4717256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/16/2018] [Accepted: 08/01/2018] [Indexed: 12/02/2022] Open
Abstract
Cerebral hyperperfusion syndrome (CHPS) is a complication that can occur after cerebral revascularization surgeries such as superficial temporal artery- (STA-) middle cerebral artery (MCA) anastomosis, and it can lead to neurological deteriorations. CHPS is usually temporary and disappears within two weeks. The authors present a case in which speech disturbance due to CHPS lasted unexpectedly long and three months was taken for full recovery. A 40-year-old woman, with a history of medication of quetiapine, dopamine 2 receptor antagonist as an antipsychotics for depression, underwent STA-MCA anastomosis for symptomatic left MCA stenosis. On the second day after surgery, the patient exhibited mild speech disturbance which deteriorated into complete motor aphasia and persisted for one month. SPECT showed the increase of cerebral blood flow (CBF) in left cerebrum, verifying the diagnosis of CHPS. Although CBF increase disappeared one month after surgery, speech disturbance continued for additionally two months with a slow improvement. This case represents a rare clinical course of CHPS. The presumable mechanisms of the prolongation of CHPS are discussed, and the medication of quetiapine might be one possible cause by its effect on cerebral vessels as dopamine 2 receptor antagonist, posing the caution against antipsychotics in cerebrovascular surgeries.
Collapse
|
32
|
Golomb BA. Diplomats' Mystery Illness and Pulsed Radiofrequency/Microwave Radiation. Neural Comput 2018; 30:2882-2985. [PMID: 30183509 DOI: 10.1162/neco_a_01133] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Importance: A mystery illness striking U.S. and Canadian diplomats to Cuba (and now China) "has confounded the FBI, the State Department and US intelligence agencies" (Lederman, Weissenstein, & Lee, 2017). Sonic explanations for the so-called health attacks have long dominated media reports, propelled by peculiar sounds heard and auditory symptoms experienced. Sonic mediation was justly rejected by experts. We assessed whether pulsed radiofrequency/microwave radiation (RF/MW) exposure can accommodate reported facts in diplomats, including unusual ones. Observations: (1) Noises: Many diplomats heard chirping, ringing or grinding noises at night during episodes reportedly triggering health problems. Some reported that noises were localized with laser-like precision or said the sounds seemed to follow them (within the territory in which they were perceived). Pulsed RF/MW engenders just these apparent "sounds" via the Frey effect. Perceived "sounds" differ by head dimensions and pulse characteristics and can be perceived as located behind in or above the head. Ability to hear the "sounds" depends on high-frequency hearing and low ambient noise. (2) Signs/symptoms: Hearing loss and tinnitus are prominent in affected diplomats and in RF/MW-affected individuals. Each of the protean symptoms that diplomats report also affect persons reporting symptoms from RF/MW: sleep problems, headaches, and cognitive problems dominate in both groups. Sensations of pressure or vibration figure in each. Both encompass vision, balance, and speech problems and nosebleeds. Brain injury and brain swelling are reported in both. (3) Mechanisms: Oxidative stress provides a documented mechanism of RF/MW injury compatible with reported signs and symptoms; sequelae of endothelial dysfunction (yielding blood flow compromise), membrane damage, blood-brain barrier disruption, mitochondrial injury, apoptosis, and autoimmune triggering afford downstream mechanisms, of varying persistence, that merit investigation. (4) Of note, microwaving of the U.S. embassy in Moscow is historically documented. Conclusions and relevance: Reported facts appear consistent with pulsed RF/MW as the source of injury in affected diplomats. Nondiplomats citing symptoms from RF/MW, often with an inciting pulsed-RF/MW exposure, report compatible health conditions. Under the RF/MW hypothesis, lessons learned for diplomats and for RF/MW-affected civilians may each aid the other.
Collapse
|
33
|
Elmorsy E, Al-Ghafari A, Helaly ANM, Hisab AS, Oehrle B, Smith PA. Editor's Highlight: Therapeutic Concentrations of Antidepressants Inhibit Pancreatic Beta-Cell Function via Mitochondrial Complex Inhibition. Toxicol Sci 2018; 158:286-301. [PMID: 28482088 DOI: 10.1093/toxsci/kfx090] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Diabetes mellitus risk is increased by prolonged usage of antidepressants (ADs). Although various mechanisms are suggested for their diabetogenic potential, whether a direct effect of ADs on pancreatic β-cells is involved is unclear. We examined this idea for 3 ADs: paroxetine, clomipramine and, with particular emphasis, fluoxetine, on insulin secretion, mitochondrial function, cellular bioenergetics, KATP channel activity, and caspase activity in murine and human cell-line models of pancreatic β-cells. Metabolic assays showed that these ADs decreased the redox, oxidative respiration, and energetic potential of β-cells in a time and concentration dependent manner, even at a concentration of 100 nM, well within the therapeutic window. These effects were related to inhibition of mitochondrial complex I and III. Consistent with impaired mitochondrial function, lactate output was increased and insulin secretion decreased. Neither fluoxetine, antimycin nor rotenone could reactivate KATP channel activity blocked by glucose unlike the mitochondrial uncoupler, FCCP. Chronic, but not acute, AD increased oxidative stress and activated caspases, 3, 8, and 9. A close agreement was found for the rates of oxidative respiration, lactate output and modulation of KATP channel activity in MIN6 cells with those of primary murine cells; data that supports MIN6 as a valid model to study beta-cell bioenergetics. To conclude, paroxetine, clomipramine and fluoxetine were all cytotoxic at therapeutic concentrations on pancreatic beta-cells; an action suggested to arise by inhibition of mitochondrial bioenergetics, oxidative stress and induction of apoptosis. These actions help explain the diabetogenic potential of these ADs in humans.
Collapse
Affiliation(s)
- Ekramy Elmorsy
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ayat Al-Ghafari
- Biochemistry Department, Faculty of Science, King Abdulaziz University (KAU), Jeddah, Kingdom of Saudi Arabia
| | - Ahmed N M Helaly
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ahmed S Hisab
- University of Nottingham Medical School, University of Nottingham, Nottinghamshire, UK
| | - Bettina Oehrle
- University of Nottingham Medical School, University of Nottingham, Nottinghamshire, UK
| | - Paul A Smith
- University of Nottingham Medical School, University of Nottingham, Nottinghamshire, UK
| |
Collapse
|
34
|
Ishii T, Hattori K, Miyakawa T, Watanabe K, Hidese S, Sasayama D, Ota M, Teraishi T, Hori H, Yoshida S, Nunomura A, Nakagome K, Kunugi H. Increased cerebrospinal fluid complement C5 levels in major depressive disorder and schizophrenia. Biochem Biophys Res Commun 2018; 497:683-688. [PMID: 29454970 DOI: 10.1016/j.bbrc.2018.02.131] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 02/15/2018] [Indexed: 11/18/2022]
Abstract
Inflammation has been implicated in a variety of psychiatric disorders. We aimed to determine whether levels of complement C5 protein in the cerebrospinal fluid (CSF), which may reflect activation of the complement system in the brain, are altered in patients with major psychiatric disorders. Additionally, we examined possible associations of CSF C5 levels with clinical variables. Subjects comprised 89 patients with major depressive disorder (MDD), 66 patients with bipolar disorder (BPD), 96 patients with schizophrenia, and 117 healthy controls, matched for age, sex, and ethnicity (Japanese). Diagnosis was made according to the Diagnostic and Statistical Manual of Mental Disorders, 4th edition, criteria. CSF C5 levels were measured by enzyme-linked immunosorbent assay. CSF C5 levels were significantly increased in the patients with MDD (p < 0.001) and in the patients with schizophrenia (p = 0.001), compared with the healthy controls. The rate of individuals with an "abnormally high C5 level" (i.e., above the 95th percentile value of the control subjects) was significantly increased in all psychiatric groups, relative to the control group (all p < 0.01). Older age, male sex, and greater body mass index tended to associate with higher C5 levels. There was a significantly positive correlation between C5 levels and chlorpromazine-equivalent dose in the patients with schizophrenia. Thus, we found, for the first time, elevated C5 levels in the CSF of patients with major psychiatric disorders. Our results suggest that the activated complement system may contribute to neurological pathogenesis in a portion of patients with major psychiatric disorders.
Collapse
Affiliation(s)
- Takashi Ishii
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neuropsychiatry, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, Japan
| | - Kotaro Hattori
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan; Medical Genome Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Tomoko Miyakawa
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan; Medical Genome Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Kentaro Watanabe
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Shinsuke Hidese
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Daimei Sasayama
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Psychiatry, Shinshu University School of Medicine, Matsumoto, Japan
| | - Miho Ota
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Toshiya Teraishi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Hiroaki Hori
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Adult Mental Health, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Sumiko Yoshida
- Medical Genome Center, National Center of Neurology and Psychiatry, Tokyo, Japan; National Center of Neurology and Psychiatry Hospital, Tokyo, Japan
| | - Akihiko Nunomura
- Department of Neuropsychiatry, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, Japan
| | - Kazuyuki Nakagome
- National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan.
| |
Collapse
|
35
|
Pollak TA, Drndarski S, Stone JM, David AS, McGuire P, Abbott NJ. The blood-brain barrier in psychosis. Lancet Psychiatry 2018; 5:79-92. [PMID: 28781208 DOI: 10.1016/s2215-0366(17)30293-6] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 06/12/2017] [Accepted: 06/27/2017] [Indexed: 02/08/2023]
Abstract
Blood-brain barrier pathology is recognised as a central factor in the development of many neurological disorders, but much less is known about the role of the blood-brain barrier in psychiatric disorders. We review post-mortem, serum-biomarker, CSF-biomarker, and neuroimaging studies that have examined blood-brain barrier structure and function in schizophrenia and related psychoses. We consider how blood-brain barrier dysfunction could relate to glutamatergic and inflammatory abnormalities, which are increasingly understood to play a part in the pathogenesis of psychosis. Mechanisms by which the blood-brain barrier and its associated solute transporters moderate CNS availability of antipsychotic drugs are summarised. We conclude that the complex nature of blood-brain barrier dysfunction in psychosis might be relevant to many aspects of disrupted neuronal and synaptic function, increased permeability to inflammatory molecules, disrupted glutamate homoeostasis, impaired action of antipsychotics, and development of antipsychotic resistance. Future research should address the longitudinal course of blood-brain barrier alterations in psychosis, to determine whether blood-brain barrier dysfunction is a cause or consequence of the pathology associated with the disorder.
Collapse
Affiliation(s)
- Thomas A Pollak
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | | | - James M Stone
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Anthony S David
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Philip McGuire
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - N Joan Abbott
- Institute of Pharmaceutical Science, King's College London, London, UK
| |
Collapse
|
36
|
Elmorsy E, Al-Ghafari A, Aggour AM, Khan R, Amer S. The role of oxidative stress in antipsychotics induced ovarian toxicity. Toxicol In Vitro 2017; 44:190-195. [DOI: 10.1016/j.tiv.2017.07.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 05/04/2017] [Accepted: 07/12/2017] [Indexed: 01/09/2023]
|
37
|
Risperidone-Induced Renal Damage and Metabolic Side Effects: The Protective Effect of Resveratrol. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:8709521. [PMID: 28706577 PMCID: PMC5494574 DOI: 10.1155/2017/8709521] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Accepted: 04/26/2017] [Indexed: 01/31/2023]
Abstract
Objective The aim of the study was to investigate the possible protective qualities of resveratrol (RSV) against the side effects of risperidone (RIS) in an experimental model in rat kidneys with histologic and biochemical assessments. Materials and Methods Experimental procedures were performed on 35 female Sprague Dawley rats. Rats were randomly divided into five groups: control, untreated rats (n = 7) were in group 1; group 2 was given 2 mg/kg/day RIS (n = 7); group 3 was treated with 2 mg/kg/day RIS and 20 mg/kg/day RSV (n = 7); group 4 was treated with 2 mg/kg/day RIS and 40 mg/kg/day RSV (n = 7); and group 5 was treated with 2 mg/kg/day RIS and 80 mg/kg/day RSV (n = 7). All treatments were administered for two weeks by gavage. On treatment day 15, kidney tissues were removed for analysis. Results The results showed that RSV treatment reduced weight gain induced by RIS. In addition, RSV increased the total antioxidant status (TAS) and decreased serum creatinine (Cr), blood urea nitrogen (BUN), oxidative stress index (OSI), and total oxidant status (TOS) levels significantly (p < 0.05). Conclusion This study revealed that treatment with RSV might protect kidney tissues against the side effects of RIS. RSV could be an effective course of therapy to enhance therapeutic efficacy.
Collapse
|
38
|
Elmorsy E, Al-Ghafari A, Almutairi FM, Aggour AM, Carter WG. Antidepressants are cytotoxic to rat primary blood brain barrier endothelial cells at high therapeutic concentrations. Toxicol In Vitro 2017; 44:154-163. [PMID: 28712878 DOI: 10.1016/j.tiv.2017.07.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 07/10/2017] [Accepted: 07/12/2017] [Indexed: 12/20/2022]
Abstract
Antidepressants are commonly employed for the treatment of major depressive disorders and other psychiatric conditions. We investigated the relatively acute cytotoxic effects of three commonly prescribed antidepressants: fluoxetine, sertraline, and clomipramine on rat primary blood brain barrier endothelial cells over a concentration range of 0.1-100μM. At therapeutic concentrations (0.1μM) no significant cytotoxicity was observed after 4, 24, or 48h. At high therapeutic to overdose concentrations (1-100μM), antidepressants reduced cell viability in proportion to their concentration and exposure duration. At 1μM, antidepressants significantly reduced mitochondrial membrane potential. At drug concentrations producing ~50% inhibition of cell viability, all drugs significantly reduced cellular oxygen consumption rates, activities of mitochondrial complexes I and III, and triggered a significant increase of lactate production. Fluoxetine (6.5μM) and clomipramine (5.5μM) also significantly lowered transcellular transport of albumin. The mechanism of cellular cytotoxicity was evaluated and at high concentrations all drugs significantly increased the production of reactive oxygen species, and significantly increased the activity of the pro-apoptotic caspases-3, 8, and 9. Comet assays revealed that all drugs were genotoxic. Pre-incubation of cells with glutathione significantly ameliorated antidepressant-induced cytotoxicity, indicating the potential benefit of treatment of overdosed patients with antioxidants.
Collapse
Affiliation(s)
- Ekramy Elmorsy
- Departments of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Egypt; Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Ayat Al-Ghafari
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Fahd M Almutairi
- Department of Biochemistry, Faculty of Science, University of Tabuk, Saudi Arabia.
| | | | - Wayne G Carter
- School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby, UK.
| |
Collapse
|
39
|
Olivieri M, Amata E, Vinciguerra S, Fiorito J, Giurdanella G, Drago F, Caporarello N, Prezzavento O, Arena E, Salerno L, Rescifina A, Lupo G, Anfuso CD, Marrazzo A. Antiangiogenic Effect of (±)-Haloperidol Metabolite II Valproate Ester [(±)-MRJF22] in Human Microvascular Retinal Endothelial Cells. J Med Chem 2016; 59:9960-9966. [PMID: 27739690 DOI: 10.1021/acs.jmedchem.6b01039] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
(±)-MRJF22 [(±)-2], a novel prodrug of haloperidol metabolite II (sigma-1 receptor antagonist/sigma-2 receptor agonist ligand) obtained by conjugation to valproic acid (histone deacetylase inhibitor) via an ester bond, exhibits antiangiogenic activity, being able to reduce human retinal endothelial cell (HREC) viability in a comparable manner to bevacizumab. Moreover, (±)-2 was able to significantly reduce viable cells count, endothelial cell migration, and tube formation in vascular endothelial growth factor A (VEGF-A) stimulated HREC cultures.
Collapse
Affiliation(s)
- Melania Olivieri
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Emanuele Amata
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Shila Vinciguerra
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Jole Fiorito
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Giovanni Giurdanella
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Nunzia Caporarello
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Orazio Prezzavento
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Emanuela Arena
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Loredana Salerno
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Antonio Rescifina
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Gabriella Lupo
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Carmelina Daniela Anfuso
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Agostino Marrazzo
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| |
Collapse
|
40
|
Cai Z, Zhao B, Deng Y, Shangguan S, Zhou F, Zhou W, Li X, Li Y, Chen G. Notch signaling in cerebrovascular diseases (Review). Mol Med Rep 2016; 14:2883-98. [PMID: 27574001 PMCID: PMC5042775 DOI: 10.3892/mmr.2016.5641] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 07/22/2016] [Indexed: 12/30/2022] Open
Abstract
The Notch signaling pathway is a crucial regulator of numerous fundamental cellular processes. Increasing evidence suggests that Notch signaling is involved in inflammation and oxidative stress, and thus in the progress of cerebrovascular diseases. In addition, Notch signaling in cerebrovascular diseases is associated with apoptosis, angiogenesis and the function of blood-brain barrier. Despite the contradictory results obtained to date as to whether Notch signaling is harmful or beneficial, the regulation of Notch signaling may provide a novel strategy for the treatment of cerebrovascular diseases.
Collapse
Affiliation(s)
- Zhiyou Cai
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Bin Zhao
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Yanqing Deng
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Shouqin Shangguan
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Faming Zhou
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Wenqing Zhou
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Xiaoli Li
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Yanfeng Li
- Department of Neurology, Peking Union Medical College Hospital, Beijing 100730, P.R. China
| | - Guanghui Chen
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| |
Collapse
|
41
|
Ruiz-Garcia J, Alegria-Barrero E. Cardiovascular Safety in Drug Development. J Cardiovasc Pharmacol Ther 2016; 21:507-515. [DOI: 10.1177/1074248416639719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 12/30/2015] [Indexed: 01/02/2023]
Abstract
As drug development becomes a long and demanding process, it might also become a barrier to medical progress. Drug safety concerns are responsible for many of the resources consumed in launching a new drug. Despite the money and time expended on it, a significant number of drugs are withdrawn years or decades after being in the market. Cardiovascular toxicity is one of the major reasons for those late withdrawals, meaning that many patients are exposed to unexpected serious cardiovascular risks. It seems that current methods to assess cardiovascular safety are imperfect, so new approaches to avoid the exposure to those undesirable effects are quite necessary. Endothelial dysfunction is the earliest detectable pathophysiological abnormality, which leads to the development of atherosclerosis, and it is also an independent predictor for major cardiovascular events. Endothelial toxicity might be the culprit of the cardiovascular adverse effects observed with a significant number of drugs. In this article, we suggest the regular inclusion of the best validated and less invasive endothelial function tests in the clinical phases of drug development in order to facilitate the development of drugs with safer cardiovascular profiles.
Collapse
Affiliation(s)
- Juan Ruiz-Garcia
- Department of Cardiology, Hospital Universitario de Torrejon, Madrid, Spain
- Facultad de Ciencias Biosanitarias, Universidad Francisco de Vitoria, Madrid, Spain
| | - Eduardo Alegria-Barrero
- Department of Cardiology, Hospital Universitario de Torrejon, Madrid, Spain
- Facultad de Ciencias Biosanitarias, Universidad Francisco de Vitoria, Madrid, Spain
| |
Collapse
|
42
|
García-Bueno B, Gassó P, MacDowell KS, Callado LF, Mas S, Bernardo M, Lafuente A, Meana JJ, Leza JC. Evidence of activation of the Toll-like receptor-4 proinflammatory pathway in patients with schizophrenia. J Psychiatry Neurosci 2016; 41:E46-55. [PMID: 27070349 PMCID: PMC4853215 DOI: 10.1503/jpn.150195] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Alterations in the innate immune/inflammatory system may underlie the pathophysiology of schizophrenia, but we do not understand the mechanisms involved. The main agents of innate immunity are the Toll-like receptors (TLRs), which detect molecular patterns associated with damage and pathogens. The TLR first reported was TLR4, and it is still the most studied one. METHODS We aimed to describe putative modifications to the TLR4 proinflammatory pathway using 2 different strategies in 2 cohorts of patients with schizophrenia and matched controls: 1) quantification of protein and mRNA expression in postmortem prefrontal cortex samples from 30 patients with schizophrenia and 30 controls, and 2) identification of single nucleotide polymorphisms associated with the risk of schizophrenia using whole blood samples from 214 patients with schizophrenia and 216 controls. RESULTS We found evidence of alterations in the expression of the initial elements of the TLR4 signalling pathway (TLR4, Myeloid differentiation primary response gene 88 [MyD88] and nuclear factor-κ B [NF-κB]) in the PFC of patients with schizophrenia. These alterations seem to depend on the presence/absence of antipsychotic treatment at death. Moreover, a polymorphism within the MyD88 gene was significantly associated with schizophrenia risk. LIMITATIONS The use of 2 different approaches in 2 different cohorts, the lack of a complementary neuropsychiatric group, the possible confounding effects of antipsychotic treatment and suicide are the main limitations of our study. CONCLUSION The evidence from this dual approach suggests there is an altered innate immune response in patients with chronic schizophrenia in which the TLR4 proinflammatory pathway could be affected. Improved understanding of the stimuli and mechanisms responsible for this response could lead to improved schizophrenia treatment and better control of the side effects of current antipsychotics.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Juan C. Leza
- Correspondence to: J.C. Leza, Department de Pharmacology, School of Medicine, Complutense University of Madrid;
| |
Collapse
|
43
|
Haloperidol and Risperidone at high concentrations activate an in vitro inflammatory response of RAW 264.7 macrophage cells by induction of apoptosis and modification of cytokine levels. Psychopharmacology (Berl) 2016; 233:1715-23. [PMID: 26391290 DOI: 10.1007/s00213-015-4079-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 09/07/2015] [Indexed: 12/11/2022]
Abstract
Antipsychotic drugs, such as haloperidol and risperidone, are used in long-term treatment of psychiatric patients and thus increase the risk of obesity and other metabolic dysfunctions. Available evidence suggests that these drugs have pro-inflammatory effect, which contributes to the establishment of endocrine disturbances. However, results yielded by extant studies are inconsistent. Therefore, in this work, we tested the in vitro effects of different high concentrations of haloperidol and risperidone on the activation of isolated macrophages (RAW 264.7 cell line). The results indicated that macrophages were activated by both drugs. In addition, the activation involved an increase in nitric oxide levels and apoptosis events by modulation of caspases 8 and 3 levels and a decrease of the Bcl-2/BAX gene expression ratio. Cells treated with haloperidol and risperidone also presented higher concentrations of inflammatory cytokines (IL-1β, IL-6, TNFα) and low levels of IL-6 anti-inflammatory cytokine in a dose-dependent manner. Despite the limitation of cell line studies based solely on macrophages cells, we suggest that antipsychotic drugs could potentially exacerbate inflammatory processes in peripheral tissues (blood and fat). The continued activation of macrophages could contribute to the development of obesity and other endocrine disturbances caused by the use of antipsychotic drugs.
Collapse
|
44
|
Sfera A, Osorio C, Inderias L, Cummings M. The Ticking of the Epigenetic Clock: Antipsychotic Drugs in Old Age. Front Endocrinol (Lausanne) 2016; 7:122. [PMID: 27630617 PMCID: PMC5005952 DOI: 10.3389/fendo.2016.00122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 08/23/2016] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Exposed to antipsychotic drugs (APDs), older individuals with dementing illness are at risk of cerebrovascular adverse effects (CVAE), including sudden death. Transient microvascular dysfunctions are known to occur in younger persons exposed to APDs; however, they seldom progress to CVAE, suggesting that APDs alone are insufficient for engendering this untoward effect. It is, therefore, believed that a preexistent microvascular damage is necessary for CVAE to take place, but the exact nature of this lesion remains unclear. CNS small vessel disease (SVD) is a well-known age-related risk factor for strokes, dementia, and sudden death, which may constitute the initial CVAE-predisposing pathology. Therefore, we propose the two strikes CVAE paradigm, in which SVD represents the first strike, while exposure to APDs, the second. In this model, both strikes must be present for CVAE to take place, and the neuroimaging load of white matter hyperintensities may be directly proportional with the CVAE risk. To investigate this hypothesis at the molecular level, we focused on a seemingly unrelated phenomenon: both APDs and SVD were found protective against a similar repertoire of cancers and their spread to the brain (1-4). Since microRNA-29 has shown efficacy against the same malignancies and has been associated with small vessels pathology, we narrowed our search down to this miR, hypothesizing that the APDs mechanism of action includes miR-29 upregulation, which in turn facilitates the development of SVD. AIM To assess whether miR-29 can be utilized as a peripheral blood biomarker for SVD and CVAE risk. METHOD We conducted a search of experimentally verified miR-29 target genes utilizing the public domain tools miRanda, RNA22 and Weizemann Institute of Science miRNA Analysis. We identified in total 67 experimentally verified target genes for miR-29 family, 18 of which correlate with microvascular integrity and may be relevant for CVAE. CONCLUSION Upregulated microRNA-29 silences the expression of 18 genes connected with capillary stability, engendering a major vulnerability for SVD (first strike) which in turn increases the risk for CVAE after exposure to APDs (second strike).
Collapse
Affiliation(s)
- Adonis Sfera
- Psychiatry, Patton State Hospital, Patton, CA, USA
- *Correspondence: Adonis Sfera,
| | | | | | | |
Collapse
|
45
|
Abstract
Schizophrenia ranks among the leading causes of disability worldwide. The presence of neurological signs co-occurring with the psychiatric symptoms is indicative of an organic brain pathology. In the present article, we review the current literature on neurology issues in schizophrenia. Firstly, common neurological signs found in patients with schizophrenia (neurological soft signs and smell abnormalities) and their association with imaging findings are reviewed. Secondly, the significant association of schizophrenia with epilepsy and stroke is described as well as the absent association with other organic brain diseases such as multiple sclerosis. Thirdly, we discuss the potential role of NMDA receptor antibodies in schizophrenia. Fourthly, neurological side effects of antipsychotic drugs and their treatment are reviewed; and lastly, we discuss neurocognitive deficits in patients with schizophrenia and their treatment. The focus of the review remains on articles with relevance to the clinician.
Collapse
Affiliation(s)
- Katharina Hüfner
- Department of Psychiatry & Psychotherapy, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria,
| | | | | |
Collapse
|
46
|
Elmorsy E, Smith PA. Bioenergetic disruption of human micro-vascular endothelial cells by antipsychotics. Biochem Biophys Res Commun 2015; 460:857-62. [PMID: 25824037 DOI: 10.1016/j.bbrc.2015.03.122] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Accepted: 03/21/2015] [Indexed: 01/16/2023]
Abstract
Antipsychotics (APs) are widely used medications, however these are not without side effects such as disruption of blood brain barrier function (BBB). To investigate this further we have studied the chronic effects of the typical APs, chlorpromazine (CPZ) and haloperidol (HAL) and the atypical APs, risperidone (RIS) and clozapine (CLZ), on the bioenergetics of human micro-vascular endothelial cells (HBVECs) of the BBB. Alamar blue (AB) and ATP assays showed that these APs impair bioenergenesis in HBVECs in a concentration and time dependent manner. However since these effects were incomplete they suggest a population of cell bioenergetically heterogeneous, an idea supported by the bistable nature by which APs affected the mitochondrial transmembrane potential. CPZ, HAL and CLZ inhibited the activity of mitochondrial complexes I and III. Our data demonstrates that at therapeutic concentrations, APs can impair the bioenergetic status of HBVECs, an action that help explains the adverse side effects of these drugs when used clinically.
Collapse
Affiliation(s)
- Ekramy Elmorsy
- School of Life Science, University of Nottingham Medical School, Queens Medical Centre, Nottinghamshire, NG7 2UH, UK; Departments of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Egypt
| | - Paul A Smith
- School of Life Science, University of Nottingham Medical School, Queens Medical Centre, Nottinghamshire, NG7 2UH, UK.
| |
Collapse
|