1
|
Zhou R, Zhang T, Sun B. Single-Cell Transcriptional Profiling Reveals Cell Type-Specific Sex-Dependent Molecular Patterns of Schizophrenia. Int J Mol Sci 2025; 26:2227. [PMID: 40076849 PMCID: PMC11900070 DOI: 10.3390/ijms26052227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Schizophrenia (SCZ) is a debilitating psychiatric disorder marked by alterations in cognition and social behavior, resulting in profound impacts on individuals and society. Although sex-dependent disparities in the epidemiology of SCZ are well established, the biological molecular basis of these disparities remains poorly understood. Investigating cell type-specific transcriptomic profiles is critical for identifying regulatory components underlying sex-dependent molecular dysregulation in SCZ, which could serve as targets for sex-specific therapeutic interventions. To address this, we systematically analyzed publicly available single-nucleus RNA sequencing datasets to characterize cell type-specific sex-dependent gene expression profiles in the prefrontal cortex of SCZ cases. Functional enrichment analyses revealed sex-dependent dysregulation patterns of SCZ at the pathway level. Furthermore, we constructed cell type-specific gene regulatory networks for males and females, identifying SCZ-associated transcription factors that interact with sex hormones and their receptors. By incorporating drug screening results from the Connectivity Map, we established disease-gene-drug connections, elucidating sex-dependent molecular mechanisms of SCZ from the single-gene to the regulatory network level. Our findings delineate the molecular patterns of sex-dependent disparities in SCZ, uncover regulatory mechanisms driving SCZ-associated sex-dependent dysregulation, and illustrate the signal flow through which the biological sex influences downstream cellular pathways in SCZ cases. Our study provides significant evidence supporting the neuroprotective role of estrogen in the pathophysiology of female SCZ cases, while also establishing a robust foundation for the development of sex-specific therapeutic approaches for both sexes.
Collapse
Affiliation(s)
| | | | - Baofa Sun
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
2
|
Kraft J, Braun A, Awasthi S, Panagiotaropoulou G, Schipper M, Bell N, Posthuma D, Pardiñas AF, Ripke S, Heilbron K. Identifying drug targets for schizophrenia through gene prioritization. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.15.24307423. [PMID: 38798390 PMCID: PMC11118622 DOI: 10.1101/2024.05.15.24307423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Background Schizophrenia genome-wide association studies (GWASes) have identified >250 significant loci and prioritized >100 disease-related genes. However, gene prioritization efforts have mostly been restricted to locus-based methods that ignore information from the rest of the genome. Methods To more accurately characterize genes involved in schizophrenia etiology, we applied a combination of highly-predictive tools to a published GWAS of 67,390 schizophrenia cases and 94,015 controls. We combined both locus-based methods (fine-mapped coding variants, distance to GWAS signals) and genome-wide methods (PoPS, MAGMA, ultra-rare coding variant burden tests). To validate our findings, we compared them with previous prioritization efforts, known neurodevelopmental genes, and results from the PsyOPS tool. Results We prioritized 62 schizophrenia genes, 41 of which were also highlighted by our validation methods. In addition to DRD2, the principal target of antipsychotics, we prioritized 9 genes that are targeted by approved or investigational drugs. These included drugs targeting glutamatergic receptors (GRIN2A and GRM3), calcium channels (CACNA1C and CACNB2), and GABAB receptor (GABBR2). These also included genes in loci that are shared with an addiction GWAS (e.g. PDE4B and VRK2). Conclusions We curated a high-quality list of 62 genes that likely play a role in the development of schizophrenia. Developing or repurposing drugs that target these genes may lead to a new generation of schizophrenia therapies. Rodent models of addiction more closely resemble the human disorder than rodent models of schizophrenia. As such, genes prioritized for both disorders could be explored in rodent addiction models, potentially facilitating drug development.
Collapse
Affiliation(s)
- Julia Kraft
- Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Alice Braun
- Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Swapnil Awasthi
- Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Georgia Panagiotaropoulou
- Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | | | - Nathaniel Bell
- Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Danielle Posthuma
- Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Child and Adolescent Psychiatry and Pediatric Psychology, Section Complex Trait Genetics, Amsterdam Neuroscience, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Antonio F. Pardiñas
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | | | - Stephan Ripke
- Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Karl Heilbron
- Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| |
Collapse
|
3
|
Debs SR, Conn I, Navaneethan B, Penklis AG, Meyer U, Killcross S, Weickert CS, Purves-Tyson TD. Maternal immune activation and estrogen receptor modulation induce sex-specific dopamine-related behavioural and molecular alterations in adult rat offspring. Brain Behav Immun 2024; 118:236-251. [PMID: 38431238 DOI: 10.1016/j.bbi.2024.02.034] [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: 11/24/2023] [Revised: 02/08/2024] [Accepted: 02/27/2024] [Indexed: 03/05/2024] Open
Abstract
Dopamine dysregulation contributes to psychosis and cognitive deficits in schizophrenia that can be modelled in rodents by inducing maternal immune activation (MIA). The selective estrogen receptor (ER) modulator, raloxifene, can improve psychosis and cognition in men and women with schizophrenia. However, few studies have examined how raloxifene may exert its therapeutic effects in mammalian brain in both sexes during young adulthood (age relevant to most prevalent age at diagnosis). Here, we tested the extent to which raloxifene alters dopamine-related behaviours and brain transcripts in young adult rats, both control and MIA-exposed females and males. We found that raloxifene increased amphetamine (AMPH)-induced locomotor activity in female controls, and in contrast, raloxifene reduced AMPH-induced locomotor activity in male MIA offspring. We did not detect overt prepulse inhibition (PPI) deficits in female or male MIA offspring, yet raloxifene enhanced PPI in male MIA offspring. Whereas, raloxifene ameliorated increased startle responsivity in female MIA offspring. In the substantia nigra (SN), we found reduced Drd2s mRNA in raloxifene-treated female offspring with or without MIA, and increased Comt mRNA in placebo-treated male MIA offspring relative to placebo-treated controls. These data demonstrate an underlying dopamine dysregulation in MIA animals that can become more apparent with raloxifene treatment, and may involve selective alterations in dopamine receptor levels and dopamine breakdown processes in the SN. Our findings support sex-specific, differential behavioural responses to ER modulation in MIA compared to control offspring, with beneficial effects of raloxifene treatment on dopamine-related behaviours relevant to schizophrenia found in male MIA offspring only.
Collapse
Affiliation(s)
- Sophie R Debs
- Preclinical Neuropsychiatry Laboratory, Neuroscience Research Australia, Sydney, Australia; Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, Australia; Discipline of Psychiatry & Mental Health, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Illya Conn
- Preclinical Neuropsychiatry Laboratory, Neuroscience Research Australia, Sydney, Australia; Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, Australia
| | - Brendan Navaneethan
- Preclinical Neuropsychiatry Laboratory, Neuroscience Research Australia, Sydney, Australia
| | - Andriane G Penklis
- Preclinical Neuropsychiatry Laboratory, Neuroscience Research Australia, Sydney, Australia; Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, Australia
| | - Urs Meyer
- Institute of Pharmacology and Toxicology, University of Zürich-Vetsuisse, Zürich, Switzerland; Switzerland Neuroscience Centre Zürich, Zürich, Switzerland
| | - Simon Killcross
- School of Psychology, University of New South Wales, Sydney, Australia
| | - Cynthia Shannon Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, Australia; Discipline of Psychiatry & Mental Health, Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Neuroscience & Physiology, Upstate Medical University, Syracuse, USA
| | - Tertia D Purves-Tyson
- Preclinical Neuropsychiatry Laboratory, Neuroscience Research Australia, Sydney, Australia; Discipline of Psychiatry & Mental Health, Faculty of Medicine, University of New South Wales, Sydney, Australia.
| |
Collapse
|
4
|
Huang Z, Zhang L, Li Y, Yu Y, Shen Y, Sun X, Lou K, Luo H, Meng Z, Li H, Wei Y. Population Pharmacodynamic Models of Risperidone on PANSS Total Scores and Prolactin Levels in Schizophrenia. Pharmaceuticals (Basel) 2024; 17:148. [PMID: 38399363 PMCID: PMC10891722 DOI: 10.3390/ph17020148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/18/2024] [Accepted: 01/18/2024] [Indexed: 02/25/2024] Open
Abstract
Currently, research predominantly focuses on evaluating clinical effects at specific time points while neglecting underlying patterns within the treatment process. This study aims to analyze the dynamic alterations in PANSS total scores and prolactin levels in patients with schizophrenia treated with risperidone, along with the influencing covariates. Using data from an 8-week randomized, double-blind, multicenter clinical trial, a population pharmacodynamic model was established for the PANSS total scores of and prolactin levels in patients treated with risperidone. The base model employed was the Emax model. Covariate selection was conducted using a stepwise forward inclusion and backward elimination approach. A total of 144 patients were included in this analysis, with 807 PANSS total scores and 531 prolactin concentration values. The PANSS total scores of the patients treated with risperidone decreased over time, fitting a proportionally parameterized sigmoid Emax model with covariates including baseline score, course of the disease, gender, plasma calcium ions, and lactate dehydrogenase levels. The increase in prolactin levels conformed to the ordinary Emax model, with covariates encompassing course of the disease, gender, weight, red blood cell count, and triglyceride levels. The impacts of the baseline scores and the course of the disease on the reduction of the PANSS scores, as well as the influence of gender on the elevation of prolactin levels, each exceeded 20%. This study provides valuable quantitative data regarding PANSS total scores and prolactin levels among patients undergoing risperidone treatment across various physiological conditions.
Collapse
Affiliation(s)
- Zhiwei Huang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (Z.H.)
| | - Lei Zhang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (Z.H.)
| | - Yan Li
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (Z.H.)
| | - Yimin Yu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (Z.H.)
| | - Yifeng Shen
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (Z.H.)
| | - Xiujia Sun
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (Z.H.)
| | - Kun Lou
- CSPC Zhongqi Pharmaceutical Technology (Shijiazhuang) Co., Ltd., Shijiazhuang 050000, China
| | - Hongmei Luo
- CSPC Zhongqi Pharmaceutical Technology (Shijiazhuang) Co., Ltd., Shijiazhuang 050000, China
| | - Zhibin Meng
- CSPC Zhongqi Pharmaceutical Technology (Shijiazhuang) Co., Ltd., Shijiazhuang 050000, China
| | - Huafang Li
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (Z.H.)
- Shanghai Clinical Research Center for Mental Health, Shanghai 200030, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai 200030, China
| | - Yumei Wei
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (Z.H.)
| |
Collapse
|
5
|
Vita A, Nibbio G, Barlati S. Pharmacological Treatment of Cognitive Impairment Associated With Schizophrenia: State of the Art and Future Perspectives. SCHIZOPHRENIA BULLETIN OPEN 2024; 5:sgae013. [PMID: 39144119 PMCID: PMC11207676 DOI: 10.1093/schizbullopen/sgae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Cognitive Impairment Associated with Schizophrenia (CIAS) represents one of the core dimensions of Schizophrenia Spectrum Disorders (SSD), with an important negative impact on real-world functional outcomes of people living with SSD. Treatment of CIAS represents a therapeutic goal of considerable importance, and while cognition-oriented evidence-based psychosocial interventions are available, effective pharmacological treatment could represent a game-changer in the lives of people with SSD. The present critical review reports and discusses the evidence regarding the effects of several pharmacological agents that are available in clinical practice or are under study, commenting on both current and future perspectives of CIAS treatment. In particular, the effects on CIAS of antipsychotic medications, anticholinergic medications, benzodiazepines, which are currently commonly used in the treatment of SSD, and of iclepertin, d-serine, luvadaxistat, xanomeline-trospium, ulotaront, anti-inflammatory molecules, and oxytocin, which are undergoing regulatory trials or can be considered as experimental agents, will be reported and discussed. Currently, available pharmacological agents do not appear to provide substantial benefits on CIAS, but accurate management of antipsychotic medications and avoiding treatments that can further exacerbate CIAS represent important strategies. Some molecules that are currently being investigated in Phase 2 and Phase 3 trials have provided very promising preliminary results, but more information is currently required to assess their effectiveness in real-world contexts and to provide clear recommendations regarding their use in clinical practice. The results of ongoing and future studies will reveal whether any of these molecules represents the awaited pharmacological game-changer in the treatment of CIAS.
Collapse
Affiliation(s)
- Antonio Vita
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Department of Mental Health and Addiction Services, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Gabriele Nibbio
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Stefano Barlati
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Department of Mental Health and Addiction Services, ASST Spedali Civili of Brescia, Brescia, Italy
| |
Collapse
|
6
|
Loeffler DA. Approaches for Increasing Cerebral Efflux of Amyloid-β in Experimental Systems. J Alzheimers Dis 2024; 100:379-411. [PMID: 38875041 PMCID: PMC11307100 DOI: 10.3233/jad-240212] [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] [Accepted: 05/14/2024] [Indexed: 06/16/2024]
Abstract
Amyloid protein-β (Aβ) concentrations are increased in the brain in both early onset and late onset Alzheimer's disease (AD). In early onset AD, cerebral Aβ production is increased and its clearance is decreased, while increased Aβ burden in late onset AD is due to impaired clearance. Aβ has been the focus of AD therapeutics since development of the amyloid hypothesis, but efforts to slow AD progression by lowering brain Aβ failed until phase 3 trials with the monoclonal antibodies lecanemab and donanemab. In addition to promoting phagocytic clearance of Aβ, antibodies lower cerebral Aβ by efflux of Aβ-antibody complexes across the capillary endothelia, dissolving Aβ aggregates, and a "peripheral sink" mechanism. Although the blood-brain barrier is the main route by which soluble Aβ leaves the brain (facilitated by low-density lipoprotein receptor-related protein-1 and ATP-binding cassette sub-family B member 1), Aβ can also be removed via the blood-cerebrospinal fluid barrier, glymphatic drainage, and intramural periarterial drainage. This review discusses experimental approaches to increase cerebral Aβ efflux via these mechanisms, clinical applications of these approaches, and findings in clinical trials with these approaches in patients with AD or mild cognitive impairment. Based on negative findings in clinical trials with previous approaches targeting monomeric Aβ, increasing the cerebral efflux of soluble Aβ is unlikely to slow AD progression if used as monotherapy. But if used as an adjunct to treatment with lecanemab or donanemab, this approach might allow greater slowing of AD progression than treatment with either antibody alone.
Collapse
Affiliation(s)
- David A. Loeffler
- Department of Neurology, Beaumont Research Institute, Corewell Health, Royal Oak, MI, USA
| |
Collapse
|
7
|
Brand BA, de Boer JN, Marcelis MC, Grootens KP, Luykx JJ, Sommer IE. The Direct and Long-Term Effects of Raloxifene as Adjunctive Treatment for Schizophrenia-Spectrum Disorders: A Double-Blind, Randomized Clinical Trial. Schizophr Bull 2023; 49:1579-1590. [PMID: 37116866 PMCID: PMC10686337 DOI: 10.1093/schbul/sbad058] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
BACKGROUND AND HYPOTHESIS Several studies suggest that raloxifene, a selective estrogen receptor modulator, improves symptoms and cognition in post-menopausal women with Schizophrenia-Spectrum Disorders (SSD). We aimed to assess the effects of adjunctive raloxifene in women and men with SSD. STUDY DESIGN This parallel, randomized, double-blind, placebo-controlled trial included adult SSD patients across the Netherlands and Belgium. Participants were stratified by age, sex, and global functioning and randomly assigned 1:1 to 12-week add-on raloxifene or placebo. Primary outcomes were symptom severity at 6, 12, and 38 weeks and cognition at 12 and 38 weeks, as measured with the Positive and Negative Syndrome Scale and the Brief Assessment of Cognition in Schizophrenia, respectively. Intention-to-treat analyses were performed using linear mixed-effect models. STUDY RESULTS We assessed 261 patients for eligibility, of which 102 (28% female) were assigned to raloxifene (n = 52) or placebo (n = 48). Although we found no main effect of raloxifene, secondary sex-specific analysis showed that in women, raloxifene had beneficial effects on negative symptoms at week 6 (LSM -2.92; adjusted P = 0.020) and week 12 (LSM -3.12; adjusted P = 0.030), and on working memory at week 38 (LSM 0.73; adjusted P = 0.040), while having negative effects on working memory at week 38 in men (LSM -0.53; adjusted P = 0.026). The number of adverse events was similar between groups. CONCLUSIONS Our results do not support the use of raloxifene in patients with SSD in general, but suggest female-specific beneficial effects of raloxifene on negative symptoms and working memory. Our findings encourage further research on sex-specific pharmacotherapeutic treatment.
Collapse
Affiliation(s)
- Bodyl A Brand
- Department of Psychiatry, UMC Utrecht Brain Center, University Medical Center Utrecht (UMCU), Utrecht University, Utrecht, The Netherlands
- Department of Biomedical Sciences and Systems, Cognitive Neurosciences, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Janna N de Boer
- Department of Psychiatry, UMC Utrecht Brain Center, University Medical Center Utrecht (UMCU), Utrecht University, Utrecht, The Netherlands
- Department of Biomedical Sciences and Systems, Cognitive Neurosciences, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Machteld C Marcelis
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- Institute for Mental Health Care Eindhoven (GGzE), Eindhoven, The Netherlands
| | - Koen P Grootens
- Reinier van Arkel Institute for Mental Health Care (RvA), ‘s Hertogenbosch, The Netherlands
- Tranzo, Tilburg School of Social and Behavioral Sciences, Tilburg University, Tilburg, The Netherlands
| | - Jurjen J Luykx
- Department of Psychiatry, UMC Utrecht Brain Center, University Medical Center Utrecht (UMCU), Utrecht University, Utrecht, The Netherlands
- Department of Psychiatry, Hospital Network Antwerp (ZNA), Antwerp, Belgium
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Outpatient Second Opinion Clinic, GGNet Mental Health, Warnsveld, The Netherlands
| | - Iris E Sommer
- Department of Psychiatry, UMC Utrecht Brain Center, University Medical Center Utrecht (UMCU), Utrecht University, Utrecht, The Netherlands
- Department of Biomedical Sciences and Systems, Cognitive Neurosciences, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| |
Collapse
|
8
|
Peng Q, Dai Z, Yin J, Lv D, Luo X, Xiong S, Yang Z, Chen G, Wei Y, Wang Y, Zhang D, Wang L, Yu D, Zhao Y, Lin D, Liao Z, Zhong Y, Lin Z, Lin J. Schizophrenia plausible protective effect of microRNA-137 is potentially related to estrogen and prolactin in female patients. Front Psychiatry 2023; 14:1187111. [PMID: 37680447 PMCID: PMC10482089 DOI: 10.3389/fpsyt.2023.1187111] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 08/03/2023] [Indexed: 09/09/2023] Open
Abstract
Background Schizophrenia (SCZ) is a serious chronic mental disorder. Our previous case-control genetic association study has shown that microRNA-137 (miR-137) may only protect females against SCZ. Since estrogen, an important female sex hormone, exerts neuroprotective effects, the relationship between estrogen and miR-137 in the pathophysiology of SCZ was further studied in this study. Methods Genotyping of single-nucleotide polymorphism rs1625579 of miR-137 gene in 1,004 SCZ patients and 896 healthy controls was conducted using the iMLDR assay. The effect of estradiol (E2) on the miR-137 expression was evaluated on the human mammary adenocarcinoma cell line (MCF-7) and the mouse hippocampal neuron cell line (HT22). The relationships between serum E2, prolactin (PRL), and peripheral blood miR-137 were investigated in 41 SCZ patients and 43 healthy controls. The miR-137 and other reference miRNAs were detected by real-time fluorescent quantitative reverse transcription-PCR. Results Based on the well-known SNP rs1625579, the distributions of protective genotypes and alleles of the miR-137 gene were not different between patients and healthy controls but were marginally significantly lower in female patients. E2 upregulated the expression of miR-137 to 2.83 and 1.81 times in MCF-7 and HT22 cells, respectively. Both serum E2 and blood miR-137 were significantly decreased or downregulated in SCZ patients, but they lacked expected positive correlations with each other in both patients and controls. When stratified by sex, blood miR-137 was negatively correlated with serum E2 in female patients. On the other hand, serum PRL was significantly increased in SCZ patients, and the female patients had the highest serum PRL level and a negative correlation between serum PRL and blood miR-137. Conclusion The plausible SCZ-protective effect of miR-137 may be female specific, of which the underlying mechanism may be that E2 upregulates the expression of miR-137. This protective mechanism may also be abrogated by elevated PRL in female patients. These preliminary findings suggest a new genetic/environmental interaction mechanism for E2/miR-137 to protect normal females against SCZ and a novel E2/PRL/miR-137-related pathophysiology of female SCZ, implying some new antipsychotic ways for female patients in future.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Zhixiong Lin
- Department of Psychiatry, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Juda Lin
- Department of Psychiatry, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
9
|
Li Z, Wang Y, Wang Z, Kong L, Liu L, Li L, Tang Y. Estradiol and raloxifene as adjunctive treatment for women with schizophrenia: A meta-analysis of randomized, double-blind, placebo-controlled trials. Acta Psychiatr Scand 2023; 147:360-372. [PMID: 36585771 DOI: 10.1111/acps.13530] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 12/13/2022] [Accepted: 12/26/2022] [Indexed: 01/01/2023]
Abstract
OBJECTIVES We conducted a comprehensive meta-analysis of all available trials to evaluate the efficacy and safety of estrogen and selective estrogen receptor modulators as adjunctive treatment for women with schizophrenia. METHODS Multiple databases were searched from the inception until March 2022. Only randomized, double-blind, placebo-controlled studies (randomized controlled trials) were included. Mean differences (MDs) and their 95% confidence intervals (CIs) were calculated using random effects models. RESULTS The meta-analysis included six estradiol versus placebo studies (n = 724) and seven raloxifene versus placebo studies (n = 419), covering a total of 1143 patients. Adjunctive estradiol outperformed the placebo in terms of the Positive and Negative Syndrome Scale (PANSS) total score (MD = -7.29; 95% CI = -10.67 to -3.91; I2 = 59.1%; p < 0.001; k = 9; N = 858), positive symptom score (MD = -1.54; 95% CI = -3.04 to -0.72; I2 = 45.8%; p < 0.001; k = 7; N = 624), negative symptom score (MD = -1.9; 95% CI = -1.77 to -0.34; I2 = 37.6%; p < 0.05; k = 14; N = 1042), and general psychopathology score (MD = -4.27; 95% CI = -7.14 to -1.41; I2 = 76.3%; p < 0.005; k = 7; N = 624). Adjunctive raloxifene outperformed the placebo in terms of the PANSS total score (MD = -6.83; 95% CI = -11.69 to -1.97; I2 = 67.8%; p = 0.006; k = 8; N = 432) and general psychopathology score (MD = -3.82; 95% CI = -6.36 to -1.28; I2 = 65.3%; p < 0.005; k = 8; N = 432). CONCLUSIONS Our meta-analysis showed that estradiol and raloxifene are effective and safe adjunctive treatments that improve schizophrenia symptoms in women. Moreover, the effects of estradiol and raloxifene differed in terms of timing and dosage. Both are promising adjunctive treatments that merit further study.
Collapse
Affiliation(s)
- Zijia Li
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yucheng Wang
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, People's Republic of China
- School of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Zhe Wang
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Lingtao Kong
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Linzi Liu
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Liu Li
- Shenyang Women's and Children's Hospital, Shenyang, People's Republic of China
| | - Yanqing Tang
- Department of Psychiatry and Geriatrics, The First Hospital of China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
10
|
Maroney M. Management of cognitive and negative symptoms in schizophrenia. Ment Health Clin 2022; 12:282-299. [DOI: 10.9740/mhc.2022.10.282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 09/08/2022] [Indexed: 11/06/2022] Open
Abstract
Abstract
Currently available antipsychotics provide only modest benefit in managing the cognitive and negative symptoms of schizophrenia even though these symptoms are often the most impairing in patients' daily lives. Certain antipsychotics may have slight benefits over others, and several nonpharmacologic and pharmacologic adjunctive treatments have been evaluated in recent clinical trials. Recently published meta-analyses and clinical studies of such treatments are reviewed. Potential strategies to manage cognitive and negative symptoms, including deprescribing of medications that may exacerbate these symptoms, are described using theoretical case examples.
Collapse
Affiliation(s)
- Megan Maroney
- 1 (Corresponding author) Clinical Associate Professor, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey; Clinical Psychiatric Pharmacist, Monmouth Medical Center, Long Branch, New Jersey,
| |
Collapse
|
11
|
Harrison PJ, Husain SM, Lee H, Los Angeles AD, Colbourne L, Mould A, Hall NAL, Haerty W, Tunbridge EM. CACNA1C (Ca V1.2) and other L-type calcium channels in the pathophysiology and treatment of psychiatric disorders: Advances from functional genomics and pharmacoepidemiology. Neuropharmacology 2022; 220:109262. [PMID: 36154842 DOI: 10.1016/j.neuropharm.2022.109262] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/09/2022] [Accepted: 09/17/2022] [Indexed: 11/17/2022]
Abstract
A role for voltage-gated calcium channels (VGCCs) in psychiatric disorders has long been postulated as part of a broader involvement of intracellular calcium signalling. However, the data were inconclusive and hard to interpret. We review three areas of research that have markedly advanced the field. First, there is now robust genomic evidence that common variants in VGCC subunit genes, notably CACNA1C which encodes the L-type calcium channel (LTCC) CaV1.2 subunit, are trans-diagnostically associated with psychiatric disorders including schizophrenia and bipolar disorder. Rare variants in these genes also contribute to the risk. Second, pharmacoepidemiological evidence supports the possibility that calcium channel blockers, which target LTCCs, might have beneficial effects on the onset or course of these disorders. This is especially true for calcium channel blockers that are brain penetrant. Third, long-range sequencing is revealing the repertoire of full-length LTCC transcript isoforms. Many novel and abundant CACNA1C isoforms have been identified in human and mouse brain, including some which are enriched compared to heart or aorta, and predicted to encode channels with differing functional and pharmacological properties. These isoforms may contribute to the molecular mechanisms of genetic association to psychiatric disorders. They may also enable development of therapeutic agents that can preferentially target brain LTCC isoforms and be of potential value for psychiatric indications.
Collapse
Affiliation(s)
- Paul J Harrison
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK; Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, OX3 7JX, UK.
| | - Syed M Husain
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK
| | - Hami Lee
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK
| | | | - Lucy Colbourne
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK; Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, OX3 7JX, UK
| | - Arne Mould
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK; Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, OX3 7JX, UK
| | - Nicola A L Hall
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK
| | - Wilfried Haerty
- Earlham Institute, Norwich Research Park, Norwich, NR4 7UZ, UK; School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Elizabeth M Tunbridge
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK; Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, OX3 7JX, UK
| |
Collapse
|
12
|
Colbourne L, Harrison PJ. Brain-penetrant calcium channel blockers are associated with a reduced incidence of neuropsychiatric disorders. Mol Psychiatry 2022; 27:3904-3912. [PMID: 35618884 PMCID: PMC9708561 DOI: 10.1038/s41380-022-01615-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 04/14/2022] [Accepted: 05/06/2022] [Indexed: 02/08/2023]
Abstract
Calcium channel blockers (CCBs) differ in their ability to penetrate into the brain. Pharmacoepidemiological studies suggest that CCBs as a class may have beneficial effects on the risks and outcomes of some psychiatric and neurological disorders. It is plausible but unknown whether this effect relates to their brain penetrance. To address this, we used the TriNetX electronic health records network to identify people prescribed a brain-penetrant CCB (BP-CCB), or those given amlodipine, a CCB with low brain penetrability. We created cohorts of patients who, prior to first CCB exposure, either had to have, or could not have had, a recorded ICD-10 diagnosis in any of the following categories: psychotic disorder; affective disorder (including bipolar disorder and major depressive disorder); anxiety disorder; substance use disorder; sleep disorder; delirium; dementia, or movement disorder. Cohort pairs were propensity score matched for age, sex, race, blood pressure, body mass index, and a range of other variables. The outcomes were the incidence of these disorders measured over a two-year exposure period. Matched cohort sizes ranged from 17,896 to 49,987. In people with no prior history of psychiatric or neurodegenerative disorder, there was a significantly lower incidence of most disorders with BP-CCBs compared to amlodipine, with risk ratios ranging from 0.64 to 0.88 and an overall risk ratio of 0.88, i.e. a risk reduction of 12%. In people who did have a prior psychiatric or neurodegenerative diagnosis, differences were much smaller, but again showed lower risks for several disorders with BP-CCBs compared to amlodipine. The differences were somewhat more marked in women and in people less than 60 years old. Results were similar when comparing BP-CCBs with verapamil and diltiazem. We also compared BP-CCBs with angiotensin receptor blockers, and found an overall risk ratio of 0.94 in favour of BP-CCBs, but with differential effects across disorders including a higher risk of psychotic disorder and dementia, but a lower risk for anxiety and sleep disorders. In some analyses, there was evidence of residual confounding even after the extensive matching, in that negative control outcomes showed a reduced incidence with BP-CCBs relative to the comparator cohort. In summary, CCBs that readily penetrate the brain are associated with a lower incidence of neuropsychiatric disorders, especially first diagnoses, compared to CCBs which do not. This may reflect their blockade of neuronal voltage-gated calcium channels. The findings encourage repurposing trials using existing BP-CCBs, and suggest that novel BP-CCBs with enhanced and more selective central actions might have greater therapeutic potential for psychiatric and neurodegenerative disorders.
Collapse
Affiliation(s)
- Lucy Colbourne
- Department of Psychiatry, University of Oxford, Oxford, OX3 7JX, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, OX3 7JX, UK
| | - Paul J Harrison
- Department of Psychiatry, University of Oxford, Oxford, OX3 7JX, UK.
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, OX3 7JX, UK.
| |
Collapse
|
13
|
Carnovale C, Perrotta C, Baldelli S, Cattaneo D, Montrasio C, Barbieri SS, Pompilio G, Vantaggiato C, Clementi E, Pozzi M. Antihypertensive drugs and brain function: mechanisms underlying therapeutically beneficial and harmful neuropsychiatric effects. Cardiovasc Res 2022; 119:647-667. [PMID: 35895876 PMCID: PMC10153433 DOI: 10.1093/cvr/cvac110] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 06/15/2022] [Accepted: 06/21/2022] [Indexed: 11/14/2022] Open
Abstract
A bidirectional relationship exists between hypertension and psychiatric disorders, including unipolar and bipolar depression, anxiety, post-traumatic stress disorder (PTSD), psychosis, schizophrenia, mania, and dementia/cognitive decline. Repurposing of antihypertensive drugs to treat mental disorders is thus being explored. A systematic knowledge of the mechanisms of action and clinical consequences of the use of antihypertensive agents on neuropsychiatric functions has not been achieved yet. In this article, we review the putative role of antihypertensive agents in psychiatric disorders, discuss the targets and mechanisms of action, and examine how and to what extent specific drug classes/molecules may trigger, worsen, or mitigate psychiatric symptoms. In addition, we review pharmacokinetics (brain penetration of drugs) and pharmacogenetics data that add important information to assess risks and benefits of antihypertensive drugs in neuropsychiatric settings. The scientific literature shows robust evidence of a positive effect of α1 blockers on PTSD symptoms, nightmares and sleep quality, α2 agonists on core symptoms, executive function and quality of life in Attention-Deficit/Hyperactivity Disorder, PTSD, Tourette's syndrome, and β blockers on anxiety, aggression, working memory, and social communication. Renin-angiotensin system modulators exert protective effects on cognition, depression, and anxiety, and the loop diuretic bumetanide reduced the core symptoms of autism in a subset of patients. There is no evidence of clear benefits of calcium channel blockers in mood disorders in the scientific literature. These findings are mainly from preclinical studies; clinical data are still insufficient or of anecdotal nature, and seldom systematic. The information herewith provided can support a better therapeutic approach to hypertension, tailored to patients with, or with high susceptibility to, psychiatric illness. It may prompt clinical studies exploring the potential benefit of antihypertensive drugs in selected patients with neuropsychiatric comorbidities that include outcomes of neuropsychiatric interest and specifically assess undesirable effects or interactions.
Collapse
Affiliation(s)
- Carla Carnovale
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences (DIBIC), ASST Fatebenefratelli-Sacco University Hospital, Università degli Studi di Milano, 20157 Milano, Italy
| | - Cristiana Perrotta
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences (DIBIC), ASST Fatebenefratelli-Sacco University Hospital, Università degli Studi di Milano, 20157 Milano, Italy
| | - Sara Baldelli
- Unit of Clinical Pharmacology, ASST Fatebenefratelli-Sacco University Hospital, 20157 Milano, Italy
| | - Dario Cattaneo
- Unit of Clinical Pharmacology, ASST Fatebenefratelli-Sacco University Hospital, 20157 Milano, Italy
| | - Cristina Montrasio
- Unit of Clinical Pharmacology, ASST Fatebenefratelli-Sacco University Hospital, 20157 Milano, Italy
| | - Silvia S Barbieri
- Unit of Brain-Heart axis: cellular and molecular mechanisms - Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine - Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy.,Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, Milan, Italy
| | | | - Emilio Clementi
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences (DIBIC), ASST Fatebenefratelli-Sacco University Hospital, Università degli Studi di Milano, 20157 Milano, Italy.,Scientific Institute IRCCS Eugenio Medea, Bosisio Parini (LC), Italy
| | - Marco Pozzi
- Scientific Institute IRCCS Eugenio Medea, Bosisio Parini (LC), Italy
| |
Collapse
|
14
|
Lanzetti S, Di Biase V. Small Molecules as Modulators of Voltage-Gated Calcium Channels in Neurological Disorders: State of the Art and Perspectives. Molecules 2022; 27:1312. [PMID: 35209100 PMCID: PMC8879281 DOI: 10.3390/molecules27041312] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/11/2022] [Accepted: 02/11/2022] [Indexed: 01/03/2023] Open
Abstract
Voltage-gated calcium channels (VGCCs) are widely expressed in the brain, heart and vessels, smooth and skeletal muscle, as well as in endocrine cells. VGCCs mediate gene transcription, synaptic and neuronal structural plasticity, muscle contraction, the release of hormones and neurotransmitters, and membrane excitability. Therefore, it is not surprising that VGCC dysfunction results in severe pathologies, such as cardiovascular conditions, neurological and psychiatric disorders, altered glycemic levels, and abnormal smooth muscle tone. The latest research findings and clinical evidence increasingly show the critical role played by VGCCs in autism spectrum disorders, Parkinson's disease, drug addiction, pain, and epilepsy. These findings outline the importance of developing selective calcium channel inhibitors and modulators to treat such prevailing conditions of the central nervous system. Several small molecules inhibiting calcium channels are currently used in clinical practice to successfully treat pain and cardiovascular conditions. However, the limited palette of molecules available and the emerging extent of VGCC pathophysiology require the development of additional drugs targeting these channels. Here, we provide an overview of the role of calcium channels in neurological disorders and discuss possible strategies to generate novel therapeutics.
Collapse
Affiliation(s)
| | - Valentina Di Biase
- Institute of Pharmacology, Department of Medical Statistics, Informatics and Health Economics, Medical University of Innsbruck, Peter-Mayr Strasse 1, A-6020 Innsbruck, Austria;
| |
Collapse
|
15
|
Pisanu C, Congiu D, Severino G, Ardau R, Chillotti C, Del Zompo M, Baune BT, Squassina A. Investigation of genetic loci shared between bipolar disorder and risk-taking propensity: potential implications for pharmacological interventions. Neuropsychopharmacology 2021; 46:1680-1692. [PMID: 34035470 PMCID: PMC8280111 DOI: 10.1038/s41386-021-01045-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 05/05/2021] [Accepted: 05/11/2021] [Indexed: 11/09/2022]
Abstract
Patients with bipolar disorder (BD) often show increased risk-taking propensity, which may contribute to poor clinical outcome. While these two phenotypes are genetically correlated, there is scarce knowledge on the shared genetic determinants. Using GWAS datasets on BD (41,917 BD cases and 371,549 controls) and risk-taking (n = 466,571), we dissected shared genetic determinants using conjunctional false discovery rate (conjFDR) and local genetic covariance analysis. We investigated specificity of identified targets using GWAS datasets on schizophrenia (SCZ) and attention-deficit hyperactivity disorder (ADHD). The putative functional role of identified targets was evaluated using different tools and GTEx v. 8. Target druggability was evaluated using DGIdb and enrichment for drug targets with genome for REPositioning drugs (GREP). Among 102 loci shared between BD and risk-taking, 87% showed the same direction of effect. Sixty-two were specifically shared between risk-taking propensity and BD, while the others were also shared between risk-taking propensity and either SCZ or ADHD. By leveraging pleiotropic enrichment, we reported 15 novel and specific loci associated with BD and 22 with risk-taking. Among cross-disorder genes, CACNA1C (a known target of calcium channel blockers) was significantly associated with risk-taking propensity and both BD and SCZ using conjFDR (p = 0.001 for both) as well as local genetic covariance analysis, and predicted to be differentially expressed in the cerebellar hemisphere in an eQTL-informed gene-based analysis (BD, Z = 7.48, p = 3.8E-14; risk-taking: Z = 4.66, p = 1.6E-06). We reported for the first time shared genetic determinants between BD and risk-taking propensity. Further investigation into calcium channel blockers or development of innovative ligands of calcium channels might form the basis for innovative pharmacotherapy in patients with BD with increased risk-taking propensity.
Collapse
Affiliation(s)
- Claudia Pisanu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Donatella Congiu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Giovanni Severino
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Raffaella Ardau
- Unit of Clinical Pharmacology of the University Hospital of Cagliari, Cagliari, Italy
| | - Caterina Chillotti
- Unit of Clinical Pharmacology of the University Hospital of Cagliari, Cagliari, Italy
| | - Maria Del Zompo
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
- Unit of Clinical Pharmacology of the University Hospital of Cagliari, Cagliari, Italy
| | - Bernhard T Baune
- Department of Psychiatry, University of Münster, Münster, Germany
- Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Alessio Squassina
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy.
| |
Collapse
|
16
|
Air Pollution-Related Brain Metal Dyshomeostasis as a Potential Risk Factor for Neurodevelopmental Disorders and Neurodegenerative Diseases. ATMOSPHERE 2020. [DOI: 10.3390/atmos11101098] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Increasing evidence links air pollution (AP) exposure to effects on the central nervous system structure and function. Particulate matter AP, especially the ultrafine (nanoparticle) components, can carry numerous metal and trace element contaminants that can reach the brain in utero and after birth. Excess brain exposure to either essential or non-essential elements can result in brain dyshomeostasis, which has been implicated in both neurodevelopmental disorders (NDDs; autism spectrum disorder, schizophrenia, and attention deficit hyperactivity disorder) and neurodegenerative diseases (NDGDs; Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, and amyotrophic lateral sclerosis). This review summarizes the current understanding of the extent to which the inhalational or intranasal instillation of metals reproduces in vivo the shared features of NDDs and NDGDs, including enlarged lateral ventricles, alterations in myelination, glutamatergic dysfunction, neuronal cell death, inflammation, microglial activation, oxidative stress, mitochondrial dysfunction, altered social behaviors, cognitive dysfunction, and impulsivity. Although evidence is limited to date, neuronal cell death, oxidative stress, and mitochondrial dysfunction are reproduced by numerous metals. Understanding the specific contribution of metals/trace elements to this neurotoxicity can guide the development of more realistic animal exposure models of human AP exposure and consequently lead to a more meaningful approach to mechanistic studies, potential intervention strategies, and regulatory requirements.
Collapse
|