1
|
Leite JSM, Vilas-Boas EA, Takahashi HK, Munhoz AC, Araújo LCC, Carvalho CR, Jr JD, Curi R, Carpinelli AR, Cruzat V. Liver lipid metabolism, oxidative stress, and inflammation in glutamine-supplemented ob/ob mice. J Nutr Biochem 2025:109842. [PMID: 39824260 DOI: 10.1016/j.jnutbio.2025.109842] [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: 06/05/2024] [Revised: 12/21/2024] [Accepted: 01/13/2025] [Indexed: 01/20/2025]
Abstract
Glutamine availability may be reduced in chronic diseases, such as type 2 diabetes mellitus (T2DM)-induced by obesity. Herein, the antioxidant, anti-inflammatory and lipid metabolism effects of chronic oral glutamine supplementation in its free and dipeptide form were assessed in ob/ob mice. Adult male C57BL/6J ob/ob mice were supplemented with L-alanyl-L-glutamine (DIP) or free L-glutamine (GLN) in the drinking water for 40 days, whilst C57BL/6J Wild-type lean (WT) and control ob/ob mice (CTRL) received fresh water only. Plasma and tissue (skeletal muscle and liver) glutamine levels, and insulin resistance parameters (e.g., GTT, ITT, insulin) were determined. Oxidative stress (e.g., GSH system, Nrf2 translocation), inflammatory (e.g., NFkB translocation, TNF-α gene expression) and lipid metabolism parameters (e.g., plasma and liver triglyceride levels, SRBP-1, FAS, ACC, and ChRBP gene expression) were also analyzed. CTRL ob/ob mice showed lower glutamine levels in plasma and tissue, as well as increased insulin resistance and fat in the liver. Conversely, chronic DIP supplementation restored glutamine levels in plasma and tissues, improved glucose homeostasis and reduced plasma and liver lipid levels. Also, Nrf2 restoration, reduced NFkB translocation, and lower TNF-α gene expression was observed in the DIP group. Interestingly, chronic free GLN only increased muscle glutamine stores but reduced overall insulin resistance, and attenuated plasma and liver lipid metabolic biomarkers. The results presented herein indicate that restoration of body glutamine levels reduces oxidative stress and inflammation in obese and T2DM ob/ob mice. This effect attenuated hepatic lipid metabolic changes observed in obesity.
Collapse
Affiliation(s)
- Jaqueline Santos Moreira Leite
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Eloisa Aparecida Vilas-Boas
- Department of Clinical and Toxicological Analysis, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Hilton K Takahashi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Ana Cláudia Munhoz
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Layanne C C Araújo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Carla Roberta Carvalho
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Jose Donato Jr
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Rui Curi
- Interdisciplinary Post-graduate Program in Health Sciences, ICAFE, Universidade Cruzeiro do Sul, São Paulo, SP, Brazil; Instituto Butantan, São Paulo, SP, Brazil
| | - Angelo Rafael Carpinelli
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Vinicius Cruzat
- Faculty of Health, Southern Cross University, Gold Coast, QLD, 4225, Australia.
| |
Collapse
|
2
|
Sethi P, Mehan S, Khan Z, Maurya PK, Kumar N, Kumar A, Tiwari A, Sharma T, Das Gupta G, Narula AS, Kalfin R. The SIRT-1/Nrf2/HO-1 axis: Guardians of neuronal health in neurological disorders. Behav Brain Res 2025; 476:115280. [PMID: 39368713 DOI: 10.1016/j.bbr.2024.115280] [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: 06/21/2024] [Revised: 09/10/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
SIRT1 (Sirtuin 1) is a NAD+-dependent deacetylase that functions through nucleoplasmic transfer and is present in nearly all mammalian tissues. SIRT1 is believed to deacetylate its protein substrates, resulting in neuroprotective actions, including reduced oxidative stress and inflammation, increased autophagy, increased nerve growth factors, and preserved neuronal integrity in aging or neurological disease. Nrf2 is a transcription factor that regulates the genes responsible for oxidative stress response and substance detoxification. The activation of Nrf2 guards cells against oxidative damage, inflammation, and carcinogenic stimuli. Several neurological abnormalities and inflammatory disorders have been associated with variations in Nrf2 activation caused by either pharmacological or genetic factors. Recent evidence indicates that Nrf2 is at the center of a complex cellular regulatory network, establishing it as a transcription factor with genuine pleiotropy. HO-1 is most likely a component of a defense mechanism in cells under stress, as it provides negative feedback for cell activation and mediator synthesis. This mediator is upregulated by Nrf2, nitric oxide (NO), and other factors in various inflammatory states. HO-1 or its metabolites, such as CO, may mitigate inflammation by modulating signal transduction pathways. Neurological diseases may be effectively treated by modulating the activity of HO-1. Multiple studies have demonstrated that SIRT1 and Nrf2 share an important connection. SIRT1 enhances Nrf2, activates HO-1, protects against oxidative injury, and decreases neuronal death. This has been associated with numerous neurodegenerative and neuropsychiatric disorders. Therefore, activating the SIRT1/Nrf2/HO-1 pathway may help treat various neurological disorders. This review focuses on the current understanding of the SIRT1 and Nrf2/HO-1 neuroprotective processes and the potential therapeutic applications of their target activators in neurodegenerative and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Pranshul Sethi
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India.
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Pankaj Kumar Maurya
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Nitish Kumar
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh 201204, India
| | - Aakash Kumar
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Aarti Tiwari
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Tarun Sharma
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| | - Reni Kalfin
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev St., Block 23, Sofia 1113, Bulgaria; Department of Healthcare, South-West University "NeofitRilski", Ivan Mihailov St. 66, Blagoevgrad 2700, Bulgaria
| |
Collapse
|
3
|
Munarriz‐Cuezva E, Meana JJ. Poly (I:C)-induced maternal immune activation generates impairment of reversal learning performance in offspring. J Neurochem 2025; 169:e16212. [PMID: 39183542 PMCID: PMC11657921 DOI: 10.1111/jnc.16212] [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: 01/29/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 08/27/2024]
Abstract
Maternal immune activation (MIA) induces a variety of behavioral and brain abnormalities in offspring of rodent models, compatible with neurodevelopmental disorders, such as schizophrenia or autism. However, it remains controversial whether MIA impairs reversal learning, a basic expression of cognitive flexibility that seems to be altered in schizophrenia. In the present study, MIA was induced by administration of a single dose of polyriboinosinic-polyribocytidylic acid (Poly (I:C) (5 mg/kg i.p.)) or saline to mouse pregnant dams in gestational day (GD) 9.5. Immune activation was monitored through changes in weight and temperature. The offspring were evaluated when they reached adulthood (8 weeks) using a touchscreen-based system to investigate the effects of Poly (I:C) on discrimination and reversal learning performance. After an initial pre-training, mice were trained to discriminate between two different stimuli, of which only one was rewarded (acquisition phase). When the correct response reached above 80% values for two consecutive days, the images were reversed (reversal phase) to assess the adaptation capacity to a changing environment. Maternal Poly (I:C) treatment did not interfere with the learning process but induced deficits in reversal learning compared to control saline animals. Thus, the accuracy in the reversal phase was lower, and Poly (I:C) animals required more sessions to complete it, suggesting impairments in cognitive flexibility. This study advances the knowledge of how MIA affects behavior, especially cognitive domains that are impaired in schizophrenia. The findings support the validity of the Poly (I:C)-based MIA model as a tool to develop pharmacological treatments targeting cognitive deficits associated with neurodevelopmental disorders.
Collapse
Affiliation(s)
- Eva Munarriz‐Cuezva
- Department of Pharmacology, Faculty of Medicine and NursingUniversity of the Basque CountryLeioaBizkaiaSpain
- Centro de Investigación Biomédica en Red de Salud MentalLeioaBizkaiaSpain
| | - Jose Javier Meana
- Department of Pharmacology, Faculty of Medicine and NursingUniversity of the Basque CountryLeioaBizkaiaSpain
- Centro de Investigación Biomédica en Red de Salud MentalLeioaBizkaiaSpain
- Biobizkaia Health Research InstituteBarakaldoBizkaiaSpain
| |
Collapse
|
4
|
Ballasch I, López-Molina L, Galán-Ganga M, Sancho-Balsells A, Rodríguez-Navarro I, Borràs-Pernas S, Rabadan MA, Chen W, Pastó-Pellicer C, Flotta F, Maoyu W, Fernández-Irigoyen J, Santamaría E, Aguilar R, Dobaño C, Egri N, Hernandez C, Alfonso M, Juan M, Alberch J, Del Toro D, Arranz B, Canals JM, Giralt A. Alterations of the IKZF1-IKZF2 tandem in immune cells of schizophrenia patients regulate associated phenotypes. J Neuroinflammation 2024; 21:326. [PMID: 39695786 DOI: 10.1186/s12974-024-03320-3] [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: 06/21/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024] Open
Abstract
Schizophrenia is a complex multifactorial disorder and increasing evidence suggests the involvement of immune dysregulations in its pathogenesis. We observed that IKZF1 and IKZF2, classic immune-related transcription factors (TFs), were both downregulated in patients' peripheral blood mononuclear cells (PBMCs) but not in their brain. We generated a new mutant mouse model with a reduction in Ikzf1 and Ikzf2 to study the impact of those changes. Such mice developed deficits in the three dimensions (positive-negative-cognitive) of schizophrenia-like phenotypes associated with alterations in structural synaptic plasticity. We then studied the secretomes of cultured PBMCs obtained from patients and identified potentially secreted molecules, which depended on IKZF1 and IKZF2 mRNA levels, and that in turn have an impact on neural synchrony, structural synaptic plasticity and schizophrenia-like symptoms in in vivo and in vitro models. Our results point out that IKZF1-IKZF2-dependent immune signals negatively impact on essential neural circuits involved in schizophrenia.
Collapse
Affiliation(s)
- Iván Ballasch
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Laura López-Molina
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Marcos Galán-Ganga
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Anna Sancho-Balsells
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Irene Rodríguez-Navarro
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Sara Borràs-Pernas
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | | | - Wanqi Chen
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Carlota Pastó-Pellicer
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Francesca Flotta
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Wang Maoyu
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Joaquín Fernández-Irigoyen
- Proteomics Platform, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra UPNA, IdiSNA, 31008, Pamplona, Spain
| | - Enrique Santamaría
- Proteomics Platform, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra UPNA, IdiSNA, 31008, Pamplona, Spain
| | - Ruth Aguilar
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Carlota Dobaño
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
- CIBER Enfermedades Infecciosas (CIBERINFEC), Barcelona, Catalonia, Spain
| | - Natalia Egri
- Servei d'Immunologia, Hospital Clinic Barcelona (HCB) - CDB, Fundació Clínic de Recerca Biomèdica - IDIBAPS, Barcelona, Spain
| | | | - Miqueu Alfonso
- Parc Sanitari Sant Joan de Déu, CIBERSAM, Barcelona, Spain
| | - Manel Juan
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Spain
- Servei d'Immunologia, Hospital Clinic Barcelona (HCB) - CDB, Fundació Clínic de Recerca Biomèdica - IDIBAPS, Barcelona, Spain
- Plataforma d'Immunoteràpia HSJD-HCB, Barcelona, Spain
| | - Jordi Alberch
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
- Production and Validation Centre of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, 08036, Barcelona, Spain
| | - Daniel Del Toro
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Belén Arranz
- Parc Sanitari Sant Joan de Déu, CIBERSAM, Barcelona, Spain
| | - Josep M Canals
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
- Production and Validation Centre of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, 08036, Barcelona, Spain
| | - Albert Giralt
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Spain.
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain.
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain.
- Production and Validation Centre of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, 08036, Barcelona, Spain.
| |
Collapse
|
5
|
Sonego AB, Prado DS, Uliana DL, Cunha TM, Grace AA, Resstel LBM. Pioglitazone attenuates behavioral and electrophysiological dysfunctions induced by two-hit model of schizophrenia in adult rodent offspring. Eur Neuropsychopharmacol 2024; 89:28-40. [PMID: 39332147 PMCID: PMC11606766 DOI: 10.1016/j.euroneuro.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 08/27/2024] [Accepted: 09/01/2024] [Indexed: 09/29/2024]
Abstract
Maternal infection and stress exposure, especially during childhood and adolescence, have been implicated as risk factors for schizophrenia. Both insults induce an exacerbated inflammatory response, which could mediate disturbance of neurodevelopmental processes and, ultimately, malfunctioning of neural systems observed in this disorder. Thus, anti-inflammatory drugs, such as PPARγ agonists, may potentially be used to prevent the development of schizophrenia. Microglia culture was prepared from the offspring of saline or poly(I:C)-injected mice. The cells were pretreated with pioglitazone and then, stimulated by LPS. Proinflammatory mediators and phagocytic activity were measured. Also, pregnant rats were injected with saline or poly(I:C) on GD17. The offspring were subjected to footshock during adolescence and subsequently injected with pioglitazone or vehicle. At adulthood, behavior and dopaminergic activity were evaluated. Pioglitazone reduced proinflammatory mediators induced by poly(I:C) microglia stimulated by LPS without affecting their decreased phagocytic activity. The PPARγ agonist also prevented the emergence of social and cognitive impairments, as well as attenuated the increased number of spontaneously active dopamine neurons in the VTA, observed in both males and females from poly(I:C) and stress group. Therefore, pioglitazone could potentially prevent the emergence of the schizophrenia-like alterations induced by the two-hit model via reduction of microglial activation.
Collapse
Affiliation(s)
- Andreza B Sonego
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 14049-900, Ribeirão Preto, SP, Brazil; Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, A210 Langley Hall, 15260, Pittsburgh, PA, USA.
| | - Douglas S Prado
- Department of Immunology, University of Pittsburgh, The Assembly Building, 15213, Pittsburgh, PA, USA
| | - Daniela L Uliana
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, A210 Langley Hall, 15260, Pittsburgh, PA, USA
| | - Thiago M Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 14049-900, Ribeirão Preto, SP, Brazil
| | - Anthony A Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, A210 Langley Hall, 15260, Pittsburgh, PA, USA
| | - Leonardo B M Resstel
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 14049-900, Ribeirão Preto, SP, Brazil
| |
Collapse
|
6
|
Chen T, Meng H, Fang N, Shi P, Chen M, Liu Q, Lv L, Li W. Age-related changes in behavior profile in male offspring of rats treated with poly I:C-induced maternal immune activation in early gestation. Animal Model Exp Med 2024; 7:914-925. [PMID: 38741390 PMCID: PMC11680485 DOI: 10.1002/ame2.12417] [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: 06/27/2023] [Accepted: 03/21/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Autism and schizophrenia are environmental risk factors associated with prenatal viral infection during pregnancy. It is still unclear whether behavior phenotypes change at different developmental stages in offspring following the activation of the maternal immune system. METHODS Sprague-Dawley rats received a single caudal vein injection of 10 mg/kg polyinosinic:polycytidylic acid (poly I:C) on gestational day 9 and the offspring were comprehensively tested for behaviors in adolescence and adulthood. RESULTS Maternal serum levels of interleukin (IL)-6, IL-1β and tumor necrosis factor-α were elevated in poly I:C-treated dams. The offspring of maternal poly I:C-induced rats showed increased anxiety, impaired social approach, and progressive impaired cognitive and sensorimotor gating function. CONCLUSION Maternal immune activation led to developmental specificity behavioral impairment in offspring.
Collapse
Affiliation(s)
- Tengfei Chen
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical UniversityXinxiangChina
- International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiangChina
| | - Huadan Meng
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical UniversityXinxiangChina
- International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiangChina
| | - Ni Fang
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical UniversityXinxiangChina
- International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiangChina
| | - Peiling Shi
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical UniversityXinxiangChina
- International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiangChina
| | - Mengxue Chen
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical UniversityXinxiangChina
- International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiangChina
| | - Qing Liu
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical UniversityXinxiangChina
- International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiangChina
| | - Luxian Lv
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical UniversityXinxiangChina
- International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiangChina
- Henan Province People's HospitalZhengzhouHenanChina
| | - Wenqiang Li
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical UniversityXinxiangChina
- International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiangChina
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental DisorderXinxiang Medical UniversityXinxiangChina
| |
Collapse
|
7
|
Fujikawa R, Yamada J, Maeda S, Iinuma KM, Moriyama G, Jinno S. Inhibition of reactive oxygen species production accompanying alternatively activated microglia by risperidone in a mouse ketamine model of schizophrenia. J Neurochem 2024; 168:2690-2709. [PMID: 38770640 DOI: 10.1111/jnc.16133] [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: 11/08/2023] [Revised: 04/17/2024] [Accepted: 05/06/2024] [Indexed: 05/22/2024]
Abstract
Recent studies have highlighted the potential involvement of reactive oxygen species (ROS) and microglia, a major source of ROS, in the pathophysiology of schizophrenia. In our study, we explored how the second-generation antipsychotic risperidone (RIS) affects ROS regulation and microglial activation in the hippocampus using a mouse ketamine (KET) model of schizophrenia. KET administration resulted in schizophrenia-like behaviors in male C57BL/6J mice, such as impaired prepulse inhibition (PPI) of the acoustic startle response and hyper-locomotion. These behaviors were mitigated by RIS. We found that the gene expression level of an enzyme responsible for ROS production (Nox2), which is primarily associated with activated microglia, was lower in KET/RIS-treated mice than in KET-treated mice. Conversely, the levels of antioxidant enzymes (Ho-1 and Gclc) were higher in KET/RIS-treated mice. The microglial density in the hippocampus was increased in KET-treated mice, which was counteracted by RIS. Hierarchical cluster analysis revealed three morphological subtypes of microglia. In control mice, most microglia were resting-ramified (type I, 89.7%). KET administration shifted the microglial composition to moderately ramified (type II, 44.4%) and hyper-ramified (type III, 25.0%). In KET/RIS-treated mice, type II decreased to 32.0%, while type III increased to 34.0%. An in vitro ROS assay showed that KET increased ROS production in dissociated hippocampal microglia, and this effect was mitigated by RIS. Furthermore, we discovered that a NOX2 inhibitor could counteract KET-induced behavioral deficits. These findings suggest that pharmacological inhibition of ROS production by RIS may play a crucial role in ameliorating schizophrenia-related symptoms. Moreover, modulating microglial activation to regulate ROS production has emerged as a novel avenue for developing innovative treatments for schizophrenia.
Collapse
Affiliation(s)
- Risako Fujikawa
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jun Yamada
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shoichiro Maeda
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kyoko M Iinuma
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | - Shozo Jinno
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
8
|
Sal-Sarria S, Conejo NM, González-Pardo H. Maternal immune activation and its multifaceted effects on learning and memory in rodent offspring: A systematic review. Neurosci Biobehav Rev 2024; 164:105844. [PMID: 39106940 DOI: 10.1016/j.neubiorev.2024.105844] [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: 05/27/2024] [Revised: 07/25/2024] [Accepted: 08/02/2024] [Indexed: 08/09/2024]
Abstract
This systematic review explored the impact of maternal immune activation (MIA) on learning and memory behavior in offspring, with a particular focus on sexual dimorphism. We analyzed 20 experimental studies involving rodent models (rats and mice) exposed to either lipopolysaccharide (LPS) or POLY I:C during gestation following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Our findings reveal that most studies report a detrimental impact of MIA on the learning and memory performance of offspring, highlighting the significant role of prenatal environmental factors in neurodevelopment. Furthermore, this review underscores the complex effects of sex, with males often exhibiting more pronounced cognitive impairment compared to females. Notably, a small subset of studies report enhanced cognitive function following MIA, suggesting complex, context-dependent outcomes of prenatal immune challenges. This review also highlights sex differences caused by the effects of MIA in terms of cytokine responses, alterations in gene expression, and differences in microglial responses as factors that contribute to the cognitive outcomes observed.
Collapse
Affiliation(s)
- Saúl Sal-Sarria
- Laboratory of Neuroscience, Department of Psychology, University of Oviedo, Oviedo, Spain; Institute of Neurosciences of the Principality of Asturias (INEUROPA), Oviedo, Spain; Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Spain.
| | - Nélida M Conejo
- Laboratory of Neuroscience, Department of Psychology, University of Oviedo, Oviedo, Spain; Institute of Neurosciences of the Principality of Asturias (INEUROPA), Oviedo, Spain; Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Spain.
| | - Héctor González-Pardo
- Laboratory of Neuroscience, Department of Psychology, University of Oviedo, Oviedo, Spain; Institute of Neurosciences of the Principality of Asturias (INEUROPA), Oviedo, Spain; Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Spain.
| |
Collapse
|
9
|
Messina A, Bella F, Maccarone G, Rodolico A, Signorelli MS. Neutrophil-Lymphocyte Ratio Values in Schizophrenia: A Comparison between Oral and Long-Acting Antipsychotic Therapies. Brain Sci 2024; 14:602. [PMID: 38928602 PMCID: PMC11201385 DOI: 10.3390/brainsci14060602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/06/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Schizophrenia is a mental disorder affecting approximately 0.32% of the global population, according to the World Health Organization. Antipsychotic medications are used to treat this condition by inhibiting D2 dopamine and 5HT2 serotonin receptors. The selection of the appropriate mode of delivery for these drugs is based on factors such as patient adherence, clinical presentation, and patient preferences. However, additional drivers of treatment selection are required in clinical practice. Mounting evidence suggests that neuroinflammation plays a crucial role in the pathogenesis of schizophrenia. NLR, a cost-effective biomarker of inflammation, has increased in several psychiatric conditions and may represent a valid method for studying the inflammatory stage in schizophrenia, relapse, and the first episode of psychosis. The aim of this study is to evaluate whether there are any variations in NLR values between patients given oral antipsychotics and those given long-acting antipsychotics. METHODS The study included 50 individuals with schizophrenia, either acute or in the follow-up phase. NLR was obtained by calculating the ratio of absolute neutrophil count (cells/μL) and absolute lymphocyte count (cells/μL). RESULTS Patients on long-acting antipsychotics exhibited significantly lower mean NLR scores (1.5 ± 0.7) compared to those on oral antipsychotics (2.2 ± 1.3) (p < 0.05). CONCLUSIONS NLR appears promising as a neuroinflammatory biomarker. This study reveals significantly lower NLR values in patients on long-acting antipsychotics, which may signify reduced systemic inflammation and improved adherence.
Collapse
Affiliation(s)
- Antonino Messina
- Psychiatry Unit, Department of Clinical and Experimental Medicine, University of Catania, 95124 Catania, Italy; (A.M.); (G.M.); (A.R.); (M.S.S.)
- Psychiatry Unit, Department of Mental Health, ASP Enna, 94100 Enna, Italy
| | - Fabrizio Bella
- Psychiatry Unit, Department of Clinical and Experimental Medicine, University of Catania, 95124 Catania, Italy; (A.M.); (G.M.); (A.R.); (M.S.S.)
| | - Giuliana Maccarone
- Psychiatry Unit, Department of Clinical and Experimental Medicine, University of Catania, 95124 Catania, Italy; (A.M.); (G.M.); (A.R.); (M.S.S.)
| | - Alessandro Rodolico
- Psychiatry Unit, Department of Clinical and Experimental Medicine, University of Catania, 95124 Catania, Italy; (A.M.); (G.M.); (A.R.); (M.S.S.)
| | - Maria Salvina Signorelli
- Psychiatry Unit, Department of Clinical and Experimental Medicine, University of Catania, 95124 Catania, Italy; (A.M.); (G.M.); (A.R.); (M.S.S.)
| |
Collapse
|
10
|
Saleki K, Alijanizadeh P, Javanmehr N, Rezaei N. The role of Toll-like receptors in neuropsychiatric disorders: Immunopathology, treatment, and management. Med Res Rev 2024; 44:1267-1325. [PMID: 38226452 DOI: 10.1002/med.22012] [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: 04/08/2022] [Revised: 10/20/2023] [Accepted: 12/20/2023] [Indexed: 01/17/2024]
Abstract
Neuropsychiatric disorders denote a broad range of illnesses involving neurology and psychiatry. These disorders include depressive disorders, anxiety, schizophrenia, bipolar disorder, attention deficit hyperactivity disorder, autism spectrum disorders, headaches, and epilepsy. In addition to their main neuropathology that lies in the central nervous system (CNS), lately, studies have highlighted the role of immunity and neuroinflammation in neuropsychiatric disorders. Toll-like receptors (TLRs) are innate receptors that act as a bridge between the innate and adaptive immune systems via adaptor proteins (e.g., MYD88) and downstream elements; TLRs are classified into 13 families that are involved in normal function and illnesses of the CNS. TLRs expression affects the course of neuropsychiatric disorders, and is influenced during their pharmacotherapy; For example, the expression of multiple TLRs is normalized during the major depressive disorder pharmacotherapy. Here, the role of TLRs in neuroimmunology, treatment, and management of neuropsychiatric disorders is discussed. We recommend longitudinal studies to comparatively assess the cell-type-specific expression of TLRs during treatment, illness progression, and remission. Also, further research should explore molecular insights into TLRs regulation and related pathways.
Collapse
Affiliation(s)
- Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- USERN Office, Babol University of Medical Sciences, Babol, Iran
- Department of e-Learning, Virtual School of Medical Education and Management, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Parsa Alijanizadeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Nima Javanmehr
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
11
|
Vargas-Calderón H, Ortega-Robles E, Rocha L, Yu P, Arias-Carrión O. Motor, Cognitive, and Behavioral Impairment in TLR3 and TLR9 Deficient Male Mice: Insights into the Non-Immunological Roles of Toll-Like Receptors. Arch Med Res 2024; 55:102985. [PMID: 38520880 DOI: 10.1016/j.arcmed.2024.102985] [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: 08/14/2023] [Revised: 02/15/2024] [Accepted: 03/05/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND Toll-like receptors (TLRs) play a critical role in initiating the innate immune response to infection or injury. Recent studies have uncovered their intriguing functions as moonlighting proteins involved in various biological processes, including development, learning, and memory. However, the specific functions of individual TLRs are still largely unknown. AIMS We investigated the effects of TLR3 and TLR9 receptor deficiency on motor, cognitive, and behavioral functions during development using genetically modified male mice of different ages. METHODS We evaluated the motor coordination, anxiety-like behavior, spatial learning, and working memory of male mice lacking the TLR3 and TLR9 genes at different ages (two, four, six, and eight months) using the rotarod, open field, water maze, and T-maze tests. RESULTS We observed that the deletion of either TLR3 or TLR9 resulted in impaired motor performance. Furthermore, young TLR3-deficient mice exhibited reduced anxiety-like behavior and spatial learning deficits; however, their working memory was unaffected. In contrast, young TLR9-knockout mice showed hyperactivity and a tendency toward decreased working memory. CONCLUSIONS These findings provide valuable insights into the broader roles of the TLR system beyond the innate immune response, revealing its involvement in pathways associated with the central nervous system. Importantly, our results establish a strong association between the endosomal receptors TLR3 and TLR9 and the performance of motor, cognitive, and behavioral tasks that change over time. This study contributes to the growing body of research on the multifaceted functions of TLRs and enhances our understanding of their participation in non-immune-related processes.
Collapse
Affiliation(s)
- Héctor Vargas-Calderón
- Unidad de Trastornos del Movimiento y Sueño, Hospital General Dr. Manuel Gea González, Ciudad de México, México; Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Emmanuel Ortega-Robles
- Unidad de Trastornos del Movimiento y Sueño, Hospital General Dr. Manuel Gea González, Ciudad de México, México
| | - Luisa Rocha
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados, Ciudad de México, México
| | - Philipp Yu
- Institut für Immunologie, Philipps-Universität Marburg, Marburg, Germany
| | - Oscar Arias-Carrión
- Unidad de Trastornos del Movimiento y Sueño, Hospital General Dr. Manuel Gea González, Ciudad de México, México.
| |
Collapse
|
12
|
Yu M, Xu J, Dutta R, Trapp B, Pieper AA, Cheng F. Network medicine informed multi-omics integration identifies drug targets and repurposable medicines for Amyotrophic Lateral Sclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.27.586949. [PMID: 38585774 PMCID: PMC10996626 DOI: 10.1101/2024.03.27.586949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a devastating, immensely complex neurodegenerative disease by lack of effective treatments. To date, the challenge to establishing effective treatment for ALS remains formidable, partly due to inadequate translation of existing human genetic findings into actionable ALS-specific pathobiology for subsequent therapeutic development. This study evaluates the feasibility of network medicine methodology via integrating human brain-specific multi-omics data to prioritize drug targets and repurposable treatments for ALS. Using human brain-specific genome-wide quantitative trait loci (x-QTLs) under a network-based deep learning framework, we identified 105 putative ALS-associated genes enriched in various known ALS pathobiological pathways, including regulation of T cell activation, monocyte differentiation, and lymphocyte proliferation. Specifically, we leveraged non-coding ALS loci effects from genome-wide associated studies (GWAS) on brain-specific expression quantitative trait loci (QTL) (eQTL), protein QTLs (pQTL), splicing QTL (sQTL), methylation QTL (meQTL), and histone acetylation QTL (haQTL). Applying network proximity analysis of predicted ALS-associated gene-coding targets and existing drug-target networks under the human protein-protein interactome (PPI) model, we identified a set of potential repurposable drugs (including Diazoxide, Gefitinib, Paliperidone, and Dimethyltryptamine) for ALS. Subsequent validation established preclinical and clinical evidence for top-prioritized repurposable drugs. In summary, we presented a network-based multi-omics framework to identify potential drug targets and repurposable treatments for ALS and other neurodegenerative disease if broadly applied.
Collapse
Affiliation(s)
- Mucen Yu
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- College of Arts and Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jielin Xu
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Ranjan Dutta
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| | - Bruce Trapp
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| | - Andrew A. Pieper
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH 44106, USA
- Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH 44106, USA
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland 44106, OH, USA
- Department of Neuroscience, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
13
|
Somayajulu M, Muhammed FS, Wright R, McClellan SA, Hazlett LD. Mechanisms of PM 10 Disruption of the Nrf2 Pathway in Cornea. Int J Mol Sci 2024; 25:3754. [PMID: 38612568 PMCID: PMC11011424 DOI: 10.3390/ijms25073754] [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: 02/13/2024] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
We have previously shown that PM10 exposure causes oxidative stress and reduces Nrf2 protein levels, and SKQ1 pre-treatment protects against this damage in human corneal epithelial cells (HCE-2). The current study focuses on uncovering the mechanisms underlying acute PM10 toxicity and SKQ1-mediated protection. HCE-2 were pre-treated with SKQ1 and then exposed to 100 μg/mL PM10. Cell viability, oxidative stress markers, programmed cell death, DNA damage, senescence markers, and pro-inflammatory cytokines were analyzed. Nrf2 cellular location and its transcriptional activity were determined. Effects of the Nrf2 inhibitor ML385 were similarly evaluated. Data showed that PM10 decreased cell viability, Nrf2 transcriptional activity, and mRNA levels of antioxidant enzymes, but increased p-PI3K, p-NFκB, COX-2, and iNOS proteins levels. Additionally, PM10 exposure significantly increased DNA damage, phosphor-p53, p16 and p21 protein levels, and β-galactosidase (β-gal) staining, which confirmed the senescence. SKQ1 pre-treatment reversed these effects. ML385 lowered the Nrf2 protein levels and mRNA levels of its downstream targets. ML385 also abrogated the protective effects of SKQ1 against PM10 toxicity by preventing the restoration of cell viability and reduced oxidative stress. In conclusion, PM10 induces inflammation, reduces Nrf2 transcriptional activity, and causes DNA damage, leading to a senescence-like phenotype, which is prevented by SKQ1.
Collapse
Affiliation(s)
| | | | | | | | - Linda D. Hazlett
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI 48201, USA; (M.S.); (F.S.M.); (R.W.); (S.A.M.)
| |
Collapse
|
14
|
Weickert TW, Ji E, Galletly C, Boerrigter D, Morishima Y, Bruggemann J, Balzan R, O’Donnell M, Liu D, Lenroot R, Weickert CS, Kindler J. Toll-Like Receptor mRNA Levels in Schizophrenia: Association With Complement Factors and Cingulate Gyrus Cortical Thinning. Schizophr Bull 2024; 50:403-417. [PMID: 38102721 PMCID: PMC10919782 DOI: 10.1093/schbul/sbad171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
BACKGROUND AND HYPOTHESES Previous studies revealed innate immune system activation in people with schizophrenia (SZ), potentially mediated by endogenous pathogen recognition receptors, notably Toll-like receptors (TLR). TLRs are activated by pathogenic molecules like bacterial lipopolysaccharides (TLR1 and TLR4), viral RNA (TLR3), or both (TLR8). Furthermore, the complement system, another key component of innate immunity, has previously been linked to SZ. STUDY DESIGN Peripheral mRNA levels of TLR1, TLR3, TLR4, and TLR8 were compared between SZ and healthy controls (HC). We investigated their relationship with immune activation through complement expression and cortical thickness of the cingulate gyrus, a region susceptible to immunological hits. TLR mRNA levels and peripheral complement receptor mRNA were extracted from 86 SZ and 77 HC white blood cells; structural MRI scans were conducted on a subset. STUDY RESULTS We found significantly higher TLR4 and TLR8 mRNA levels and lower TLR3 mRNA levels in SZ compared to HC. TLRs and complemental factors were significantly associated in SZ and HC, with the strongest deviations of TLR mRNA levels in the SZ subgroup having elevated complement expression. Cortical thickness of the cingulate gyrus was inversely associated with TLR8 mRNA levels in SZ, and with TLR4 and TLR8 levels in HC. CONCLUSIONS The study underscores the role of innate immune activation in schizophrenia, indicating a coordinated immune response of TLRs and the complement system. Our results suggest there could be more bacterial influence (based on TLR 4 levels) as opposed to viral influence (based on TLR3 levels) in schizophrenia. Specific TLRs were associated with brain cortical thickness reductions of limbic brain structures.
Collapse
Affiliation(s)
- Thomas W Weickert
- Neuroscience Research Australia, Schizophrenia Research Institute, Randwick, NSW 2031, Australia
- School of Psychiatry, University of New South Wales, Randwick, NSW 2031Australia
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY 13210, USA
| | - Ellen Ji
- Psychiatric University Hospital Zurich, Zurich, Switzerland
- Neuroscience Research Australia, Sydney, NSW, Australia
| | - Cherrie Galletly
- Discipline of Psychiatry, School of Medicine, University of Adelaide, Adelaide, SA, Australia
- Ramsay Health Care (SA) Mental Health, Adelaide, Australia
- Northern Adelaide Local Health Network, Adelaide, SA, Australia
| | - Danny Boerrigter
- Neuroscience Research Australia, Schizophrenia Research Institute, Randwick, NSW 2031, Australia
| | - Yosuke Morishima
- Translational Research Center, University Hospital of Psychiatry, University of Bern, Bern, Switzerland
| | - Jason Bruggemann
- Neuroscience Research Australia, Schizophrenia Research Institute, Randwick, NSW 2031, Australia
- School of Psychiatry, University of New South Wales, Randwick, NSW 2031Australia
- Edith Collins Centre (Translational Research in Alcohol Drugs and Toxicology), Sydney Local Health District, Sydney, Australia
- Speciality of Addiction Medicine, Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Ryan Balzan
- School of Psychology, Flinders University, Adelaide, SA, Australia
| | - Maryanne O’Donnell
- Neuroscience Research Australia, Schizophrenia Research Institute, Randwick, NSW 2031, Australia
- Kiloh Centre, Prince of Wales Hospital, Randwick, New South Wales, Australia
| | - Dennis Liu
- Discipline of Psychiatry, School of Medicine, University of Adelaide, Adelaide, SA, Australia
- Ramsay Health Care (SA) Mental Health, Adelaide, Australia
- Northern Adelaide Local Health Network, Adelaide, SA, Australia
| | - Rhoshel Lenroot
- Neuroscience Research Australia, Schizophrenia Research Institute, Randwick, NSW 2031, Australia
- School of Psychiatry, University of New South Wales, Randwick, NSW 2031Australia
- Department of Psychiatry, University of New Mexico, Albuquerque, NM 87131-0001, USA
| | - Cynthia Shannon Weickert
- Neuroscience Research Australia, Schizophrenia Research Institute, Randwick, NSW 2031, Australia
- School of Psychiatry, University of New South Wales, Randwick, NSW 2031Australia
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY 13210, USA
| | - Jochen Kindler
- Neuroscience Research Australia, Schizophrenia Research Institute, Randwick, NSW 2031, Australia
- University Hospital of Child and Adolescent Psychiatry and Psychotherapy, University of Bern, 3000 Bern, Switzerland
| |
Collapse
|
15
|
DelaCuesta-Barrutia J, Martínez-Peula O, Rivero G, Santas-Martín JA, Munarriz-Cuezva E, Brocos-Mosquera I, Miranda-Azpiazu P, Diez-Alarcia R, Morentin B, Honer WG, Callado LF, Erdozain AM, Ramos-Miguel A. Effect of antipsychotic drugs on group II metabotropic glutamate receptor expression and epigenetic control in postmortem brains of schizophrenia subjects. Transl Psychiatry 2024; 14:113. [PMID: 38396013 PMCID: PMC10891050 DOI: 10.1038/s41398-024-02832-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Antipsychotic-induced low availability of group II metabotropic glutamate receptors (including mGlu2R and mGlu3R) in brains of schizophrenia patients may explain the limited efficacy of mGlu2/3R ligands in clinical trials. Studies evaluating mGlu2/3R levels in well-designed, large postmortem brain cohorts are needed to address this issue. Postmortem samples from the dorsolateral prefrontal cortex of 96 schizophrenia subjects and matched controls were collected. Toxicological analyses identified cases who were (AP+) or were not (AP-) receiving antipsychotic treatment near the time of death. Protein and mRNA levels of mGlu2R and mGlu3R, as well as GRM2 and GRM3 promoter-attached histone posttranslational modifications, were quantified. Experimental animal models were used to compare with data obtained in human tissues. Compared to matched controls, schizophrenia cortical samples had lower mGlu2R protein amounts, regardless of antipsychotic medication. Downregulation of mGlu3R was observed in AP- schizophrenia subjects only. Greater predicted occupancy values of dopamine D2 and serotonin 5HT2A receptors correlated with higher density of mGlu3R, but not mGlu2R. Clozapine treatment and maternal immune activation in rodents mimicked the mGlu2R, but not mGlu3R regulation observed in schizophrenia brains. mGlu2R and mGlu3R mRNA levels, and the epigenetic control mechanisms did not parallel the alterations at the protein level, and in some groups correlated inversely. Insufficient cortical availability of mGlu2R and mGlu3R may be associated with schizophrenia. Antipsychotic treatment may normalize mGlu3R, but not mGlu2R protein levels. A model in which epigenetic feedback mechanisms controlling mGlu3R expression are activated to counterbalance mGluR loss of function is described.
Collapse
Affiliation(s)
| | | | - Guadalupe Rivero
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Leioa, Spain
| | - Jon A Santas-Martín
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - Eva Munarriz-Cuezva
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Leioa, Spain
| | - Iria Brocos-Mosquera
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
| | | | - Rebeca Diez-Alarcia
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Leioa, Spain
| | - Benito Morentin
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Basque Institute of Legal Medicine, Bilbao, Spain
| | - William G Honer
- Department Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Luis F Callado
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Leioa, Spain
| | - Amaia M Erdozain
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Leioa, Spain
| | - Alfredo Ramos-Miguel
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain.
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Leioa, Spain.
| |
Collapse
|
16
|
Bris ÁG, MacDowell KS, Ulecia-Morón C, Martín-Hernández D, Moreno B, Madrigal JLM, García-Bueno B, Caso JR, Leza JC. Differential regulation of innate immune system in frontal cortex and hippocampus in a "double-hit" neurodevelopmental model in rats. Neurotherapeutics 2024; 21:e00300. [PMID: 38241165 PMCID: PMC10903097 DOI: 10.1016/j.neurot.2023.10.010] [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: 10/17/2023] [Accepted: 10/28/2023] [Indexed: 01/21/2024] Open
Abstract
Neurodevelopmental disorders (NDs) are neuropsychiatric conditions affecting central nervous system development, characterized by cognitive and behavioural alterations. Inflammation has been recently linked to NDs. Animal models are essential for understanding their pathophysiology and identifying therapeutic targets. Double-hit models can reproduce neurodevelopmental and neuroinflammatory impairments. Sixty-seven newborn rats were assigned to four groups: Control, Maternal deprivation (MD, 24-h-deprivation), Isolation (Iso, 5 weeks), and Maternal deprivation + Isolation (MD + Iso, also known as double-hit). Cognitive dysfunction was assessed using behavioural tests. Inflammasome, MAPKs, and TLRs inflammatory elements expression in the frontal cortex (FC) and hippocampus (HP) was analysed through western blot and qRT-PCR. Oxidative/nitrosative (O/N) evaluation and corticosterone levels were measured in plasma samples. Double-hit group was affected in executive and working memory. Most inflammasomes and TLRs inflammatory responses were increased in FC compared to the control group, whilst MAPKs were downregulated. Conversely, hippocampal inflammasome and inflammatory components were reduced after the double-hit exposure, while MAPKs were elevated. Our findings reveal differential regulation of innate immune system components in FC and HP in the double-hit group. Further investigations on MAPKs are necessary to understand their role in regulating HP neuroinflammatory status, potentially linking our MAPKs results to cognitive impairments through their proliferative and anti-inflammatory activity.
Collapse
Affiliation(s)
- Álvaro G Bris
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Karina S MacDowell
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Cristina Ulecia-Morón
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - David Martín-Hernández
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Beatriz Moreno
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - José L M Madrigal
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Borja García-Bueno
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Javier R Caso
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Juan C Leza
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain.
| |
Collapse
|
17
|
de Oliveira EG, de Lima DA, da Silva Júnior JC, de Souza Barbosa MV, de Andrade Silva SC, de Santana JH, Dos Santos Junior OH, Lira EC, Lagranha CJ, Duarte FS, Gomes DA. (R)-ketamine attenuates neurodevelopmental disease-related phenotypes in a mouse model of maternal immune activation. Eur Arch Psychiatry Clin Neurosci 2023; 273:1501-1512. [PMID: 37249625 DOI: 10.1007/s00406-023-01629-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/22/2023] [Indexed: 05/31/2023]
Abstract
Infections during pregnancy are associated with an increased risk of neuropsychiatric disorders with developmental etiologies, such as schizophrenia and autism spectrum disorders (ASD). Studies have shown that the animal model of maternal immune activation (MIA) reproduces a wide range of phenotypes relevant to the study of neurodevelopmental disorders. Emerging evidence shows that (R)-ketamine attenuates behavioral, cellular, and molecular changes observed in animal models of neuropsychiatric disorders. Here, we investigate whether (R)-ketamine administration during adolescence attenuates some of the phenotypes related to neurodevelopmental disorders in an animal model of MIA. For MIA, pregnant Swiss mice received intraperitoneally (i.p.) lipopolysaccharide (LPS; 100 µg/kg/day) or saline on gestational days 15 and 16. The two MIA-based groups of male offspring received (R)-ketamine (20 mg/kg/day; i.p.) or saline from postnatal day (PND) 36 to 50. At PND 62, the animals were examined for anxiety-like behavior and locomotor activity in the open-field test (OFT), as well as in the social interaction test (SIT). At PND 63, the prefrontal cortex (PFC) was collected for analysis of oxidative balance and gene expression of the cytokines IL-1β, IL-6, and TGF-β1. We show that (R)-ketamine abolishes anxiety-related behavior and social interaction deficits induced by MIA. Additionally, (R)-ketamine attenuated the increase in lipid peroxidation and the cytokines in the PFC of the offspring exposed to MIA. The present work suggests that (R)-ketamine administration may have a long-lasting attenuation in deficits in emotional behavior induced by MIA, and that these effects may be attributed to its antioxidant and anti-inflammatory activity in the PFC.
Collapse
Affiliation(s)
- Elifrances Galdino de Oliveira
- Laboratory of Neuroendocrinology and Metabolism, Department of Physiology and Pharmacology, Bioscience Center, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235 Cidade Universitária, Recife, PE, 50670-901, Brazil.
- Graduate Program of Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, Recife, PE, Brazil.
| | - Diógenes Afonso de Lima
- Laboratory of Neuroendocrinology and Metabolism, Department of Physiology and Pharmacology, Bioscience Center, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235 Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - José Carlos da Silva Júnior
- Laboratory of Neuroendocrinology and Metabolism, Department of Physiology and Pharmacology, Bioscience Center, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235 Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Mayara Victória de Souza Barbosa
- Laboratory of Neuroendocrinology and Metabolism, Department of Physiology and Pharmacology, Bioscience Center, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235 Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Severina Cassia de Andrade Silva
- Graduate Program of Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, Recife, PE, Brazil
- Laboratory of Biochemistry and Exercise Biochemistry, Department of Physical Education and Sports Science, Federal University of Pernambuco, Vitória de Santo Antão, PE, Brazil
| | - Jonata Henrique de Santana
- Graduate Program of Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, Recife, PE, Brazil
- Laboratory of Biochemistry and Exercise Biochemistry, Department of Physical Education and Sports Science, Federal University of Pernambuco, Vitória de Santo Antão, PE, Brazil
| | - Osmar Henrique Dos Santos Junior
- Graduate Program of Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, Recife, PE, Brazil
- Laboratory of Biochemistry and Exercise Biochemistry, Department of Physical Education and Sports Science, Federal University of Pernambuco, Vitória de Santo Antão, PE, Brazil
| | - Eduardo Carvalho Lira
- Laboratory of Neuroendocrinology and Metabolism, Department of Physiology and Pharmacology, Bioscience Center, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235 Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Claudia Jacques Lagranha
- Graduate Program of Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, Recife, PE, Brazil
- Laboratory of Biochemistry and Exercise Biochemistry, Department of Physical Education and Sports Science, Federal University of Pernambuco, Vitória de Santo Antão, PE, Brazil
| | - Filipe Silveira Duarte
- Laboratory of Neuroendocrinology and Metabolism, Department of Physiology and Pharmacology, Bioscience Center, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235 Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Dayane Aparecida Gomes
- Laboratory of Neuroendocrinology and Metabolism, Department of Physiology and Pharmacology, Bioscience Center, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235 Cidade Universitária, Recife, PE, 50670-901, Brazil
- Graduate Program of Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, Recife, PE, Brazil
| |
Collapse
|
18
|
Airapetov MI, Eresko SO, Ignatova PD, Skabelkin DA, Mikhailova AA, Ganshina DA, Lebedev AA, Bychkov ER, Shabanov PD. The effect of rifampicin on expression of the toll-like receptor system genes in the forebrain cortex of rats prenatally exposed to alcohol. BIOMEDITSINSKAIA KHIMIIA 2023; 69:228-234. [PMID: 37705483 DOI: 10.18097/pbmc20236904228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Ethanol causes long-term changes in the toll-like receptor (TLR) system, promoting activation of neuroinflammation pathways. Alcohol use during pregnancy causes neuroinflammatory processes in the fetus; this can lead to the development of symptoms of fetal alcohol spectrum disorder (FASD). Our study has shown that prenatal alcohol exposure (PAE) induced long-term changes in the TLR system genes (Tlr3, Tlr4, Ticam, Hmgb1, cytokine genes) in the forebrain cortex of rat pups. Administration of rifampicin (Rif), which can reduce the level of pro-inflammatory mediators in various pathological conditions of the nervous system, normalized the altered expression level of the studied TLR system genes. This suggests that Rif can prevent the development of persistent neuroinflammatory events in the forebrain cortex of rat pups caused by dysregulation in the TLR system.
Collapse
Affiliation(s)
- M I Airapetov
- Institute of Experimental Medicine, St. Petersburg, Russia; Military Medical Academy of S.M. Kirov, St. Petersburg, Russia
| | - S O Eresko
- Institute of Experimental Medicine, St. Petersburg, Russia; North-Western State Medical University named after I.I. Mechnikov, St. Petersburg, Russia
| | - P D Ignatova
- Institute of Experimental Medicine, St. Petersburg, Russia
| | - D A Skabelkin
- Institute of Experimental Medicine, St. Petersburg, Russia
| | - A A Mikhailova
- Institute of Experimental Medicine, St. Petersburg, Russia
| | - D A Ganshina
- Institute of Experimental Medicine, St. Petersburg, Russia
| | - A A Lebedev
- Institute of Experimental Medicine, St. Petersburg, Russia
| | - E R Bychkov
- Institute of Experimental Medicine, St. Petersburg, Russia
| | - P D Shabanov
- Institute of Experimental Medicine, St. Petersburg, Russia
| |
Collapse
|
19
|
Karanikas E. The Gordian knot of the immune-redox systems' interactions in psychosis. Int Clin Psychopharmacol 2023; 38:285-296. [PMID: 37351570 DOI: 10.1097/yic.0000000000000481] [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] [Indexed: 06/24/2023]
Abstract
During the last decades the attempt to enlighten the pathobiological substrate of psychosis, from merely focusing on neurotransmitters, has expanded into new areas like the immune and redox systems. Indeed, the inflammatory hypothesis concerning psychosis etiopathology has exponentially grown with findings reflecting dysfunction/aberration of the immune/redox systems' effector components namely cytokines, chemokines, CRP, complement system, antibodies, pro-/anti-oxidants, oxidative stress byproducts just to name a few. Yet, we still lie far from comprehending the underlying cellular mechanisms, their causality directions, and the moderating/mediating parameters affecting these systems; let alone the inter-systemic (between immune and redox) interactions. Findings from preclinical studies on the stress field have provided evidence indicative of multifaceted interactions among the immune and redox components so tightly intertwined as a Gordian knot. Interestingly the literature concerning the interactions between these same systems in the context of psychosis appears minimal (if not absent) and ambiguous. This review attempts to draw a frame of the immune-redox systems' interactions starting from basic research on the stress field and expanding on clinical studies with cohorts with psychosis, hoping to instigate new avenues of research.
Collapse
Affiliation(s)
- Evangelos Karanikas
- Department of Psychiatry, 424 General Military Hospital, Ring Road, Nea Efkarpia, Thessaloniki, Greece
| |
Collapse
|
20
|
Patlola SR, Donohoe G, McKernan DP. Counting the Toll of Inflammation on Schizophrenia-A Potential Role for Toll-like Receptors. Biomolecules 2023; 13:1188. [PMID: 37627253 PMCID: PMC10452856 DOI: 10.3390/biom13081188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/25/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Toll-like receptors (TLRs) are a family of pattern recognition receptors (PRRs) that are ubiquitously expressed in the human body. They protect the brain and central nervous system from self and foreign antigens/pathogens. The immune response elicited by these receptors culminates in the release of cytokines, chemokines, and interferons causing an inflammatory response, which can be both beneficial and harmful to neurodevelopment. In addition, the detrimental effects of TLR activation have been implicated in multiple neurodegenerative diseases such as Alzheimer's, multiple sclerosis, etc. Many studies also support the theory that cytokine imbalance may be involved in schizophrenia, and a vast amount of literature showcases the deleterious effects of this imbalance on cognitive performance in the human population. In this review, we examine the current literature on TLRs, their potential role in the pathogenesis of schizophrenia, factors affecting TLR activity that contribute towards the risk of schizophrenia, and lastly, the role of TLRs and their impact on cognitive performance in schizophrenia.
Collapse
Affiliation(s)
- Saahithh Redddi Patlola
- Department of Pharmacology & Therapeutics, School of Medicine, University of Galway, H91 TK33 Galway, Ireland;
| | - Gary Donohoe
- School of Psychology, University of Galway, H91 TK33 Galway, Ireland;
| | - Declan P. McKernan
- Department of Pharmacology & Therapeutics, School of Medicine, University of Galway, H91 TK33 Galway, Ireland;
| |
Collapse
|
21
|
C de S Ribeiro B, V de C Faria R, de S Nogueira J, Santos Valença S, Chen L, Romana-Souza B. Olive oil promotes the survival and migration of dermal fibroblasts through Nrf2 pathway activation. Lipids 2023; 58:59-68. [PMID: 36289559 DOI: 10.1002/lipd.12363] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/27/2022] [Accepted: 10/15/2022] [Indexed: 11/07/2022]
Abstract
Olive oil has beneficial effects on skin wound healing due to its anti-inflammatory and antioxidant properties; however, the mechanism by which olive oil promotes wound healing is unclear. We evaluated the mechanisms involved in Nrf2 pathway activation by olive oil and its role in cell survival and migration in mouse dermal fibroblasts in a short-term exposition. Our data demonstrated that olive oil and oleic acid promoted reactive oxygen species (ROS) production, while olive oil and hydroxytyrosol stimulated nuclear factor erythroid 2-related factor 2 (Nrf2) activation. Olive oil-mediated ROS production increased nuclear factor kappa B p65 expression, while olive oil-stimulated reactive nitrogen species production augmented the levels of Nrf2. Olive oil augmented cell proliferation, cell migration, and AKT phosphorylation, but decreased apoptotic cell number and cleaved caspase-3 levels. The effect of olive oil on cell migration and protein levels of AKT, BCL-2, and Nrf2 were reversed by an Nrf2 inhibitor. In conclusion, the activation of the Nrf2 pathway by olive oil promotes the survival and migration of dermal fibroblasts that are essential for the resolution of skin wound healing.
Collapse
Affiliation(s)
- Bianca C de S Ribeiro
- Department of Histology and Embryology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Regina V de C Faria
- Department of Histology and Embryology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Jeane de S Nogueira
- Histocompatibility and Cryopreservation Laboratory, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Samuel Santos Valença
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lin Chen
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, Illinois, USA
| | - Bruna Romana-Souza
- Department of Histology and Embryology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| |
Collapse
|
22
|
Enrico P, Delvecchio G, Turtulici N, Aronica R, Pigoni A, Squarcina L, Villa FM, Perlini C, Rossetti MG, Bellani M, Lasalvia A, Bonetto C, Scocco P, D'Agostino A, Torresani S, Imbesi M, Bellini F, Veronese A, Bocchio-Chiavetto L, Gennarelli M, Balestrieri M, Colombo GI, Finardi A, Ruggeri M, Furlan R, Brambilla P. A machine learning approach on whole blood immunomarkers to identify an inflammation-associated psychosis onset subgroup. Mol Psychiatry 2023; 28:1190-1200. [PMID: 36604602 DOI: 10.1038/s41380-022-01911-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 11/29/2022] [Accepted: 12/07/2022] [Indexed: 01/07/2023]
Abstract
Psychosis onset is a transdiagnostic event that leads to a range of psychiatric disorders, which are currently diagnosed through clinical observation. The integration of multimodal biological data could reveal different subtypes of psychosis onset to target for the personalization of care. In this study, we tested the existence of subgroups of patients affected by first-episode psychosis (FEP) with a possible immunopathogenic basis. To do this, we designed a data-driven unsupervised machine learning model to cluster a sample of 127 FEP patients and 117 healthy controls (HC), based on the peripheral blood expression levels of 12 psychosis-related immune gene transcripts. To validate the model, we applied a resampling strategy based on the half-splitting of the total sample with random allocation of the cases. Further, we performed a post-hoc univariate analysis to verify the clinical, cognitive, and structural brain correlates of the subgroups identified. The model identified and validated two distinct clusters: 1) a FEP cluster characterized by the high expression of inflammatory and immune-activating genes (IL1B, CCR7, IL12A and CXCR3); 2) a cluster consisting of an equal number of FEP and HC subjects, which did not show a relative over or under expression of any immune marker (balanced subgroup). None of the subgroups was related to specific symptoms dimensions or longitudinal diagnosis of affective vs non-affective psychosis. FEP patients included in the balanced immune subgroup showed a thinning of the left supramarginal and superiorfrontal cortex (FDR-adjusted p-values < 0.05). Our results demonstrated the existence of a FEP patients' subgroup identified by a multivariate pattern of immunomarkers involved in inflammatory activation. This evidence may pave the way to sample stratification in clinical studies aiming to develop diagnostic tools and therapies targeting specific immunopathogenic pathways of psychosis.
Collapse
Affiliation(s)
- Paolo Enrico
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.,Department of Psychiatry and Psychotherapy, Ludwig-Maximilian-University, Munich, Germany
| | - Giuseppe Delvecchio
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Nunzio Turtulici
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Rosario Aronica
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Alessandro Pigoni
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,Social and Affective Neuroscience Group, MoMiLab, IMT School for Advanced Studies Lucca, Lucca, Italy
| | - Letizia Squarcina
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Filippo M Villa
- Child Psychopathology Unit, Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Lecco, Italy
| | - Cinzia Perlini
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Clinical Psychology, University of Verona, Verona, Italy.,USD Clinical Psychology, Azienda Ospedaliera Universitaria Integrata (AOUI) of Verona, Verona, Italy
| | - Maria G Rossetti
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Marcella Bellani
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Psychiatry, University of Verona, Verona, Italy.,UOC of Psychiatry, Azienda Ospedaliera Universitaria Integrata (AOUI) of Verona, Verona, Italy
| | - Antonio Lasalvia
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Psychiatry, University of Verona, Verona, Italy
| | - Chiara Bonetto
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Psychiatry, University of Verona, Verona, Italy
| | - Paolo Scocco
- Department of Mental Health, AULSS 6 Euganea, Padua, Italy
| | - Armando D'Agostino
- Department of Health Sciences, San Paolo University Hospital, University of Milan, Milano, Milan, Italy
| | - Stefano Torresani
- Department of Psychiatry, ULSS, Bolzano Suedtiroler Sanitaetbetrieb- Azienda Sanitaria dell'Alto Adige, Bolzano, Italy
| | | | | | | | - Luisella Bocchio-Chiavetto
- Faculty of Psychology, eCampus University, Novedrate, Como, Italy.,Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio, Fatebenefratelli, Brescia, Italy
| | - Massimo Gennarelli
- Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio, Fatebenefratelli, Brescia, Italy.,Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | - Gualtiero I Colombo
- Centro Cardiologico Monzino IRCCS, Immunology and Functional Genomics Unit, Milan, Italy
| | - Annamaria Finardi
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Mirella Ruggeri
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Psychiatry, University of Verona, Verona, Italy.,UOC of Psychiatry, Azienda Ospedaliera Universitaria Integrata (AOUI) of Verona, Verona, Italy
| | - Roberto Furlan
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Paolo Brambilla
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy. .,Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.
| | | |
Collapse
|
23
|
Karanikas E. The immune-stress/endocrine-redox-metabolic nature of psychosis' etiopathology; focus on the intersystemic pathways interactions. Neurosci Lett 2023; 794:137011. [PMID: 36513162 DOI: 10.1016/j.neulet.2022.137011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/26/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
Abstract
The evidence supporting the involvement of a number of systems in the neurobiological etiopathology of psychosis has recently grown exponentially. Indeed, the focus of research has changed from measuring solely neurotransmitters to estimating parameters from fields like immunity, stress/endocrine, redox, and metabolism. Yet, little is known regarding the exact role of each one of these fields on the formation of not only the brain neuropathological substrate in psychosis but also the associated general systemic pathology, in terms of causality directions. Research has shown deviations in the levels and/or function of basic effector molecules of the aforementioned fields namely cytokines, pro-/anti- oxidants, glucocorticoids, catecholamines, glucose, and lipids metabolites as well as kynurenines, in psychosis. Yet the evidence regarding their impact on neurotransmitters is minimal and the findings concerning these systems' interactions in the psychotic context are even more dispersed. The present review aims to draw holistically the frame of the hitherto known "players" in the field of psychosis' cellular pathobiology, with a particular focus on their in-between interactions.
Collapse
Affiliation(s)
- Evangelos Karanikas
- Department of Psychiatry, 424 General Military Hospital, Thessaloniki, Greece.
| |
Collapse
|
24
|
Li J, Wang Y, Yuan X, Kang Y, Song X. New insight in the cross-talk between microglia and schizophrenia: From the perspective of neurodevelopment. Front Psychiatry 2023; 14:1126632. [PMID: 36873215 PMCID: PMC9978517 DOI: 10.3389/fpsyt.2023.1126632] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 01/27/2023] [Indexed: 02/18/2023] Open
Abstract
Characterized by psychotic symptoms, negative symptoms and cognitive deficits, schizophrenia had a catastrophic effect on patients and their families. Multifaceted reliable evidence indicated that schizophrenia is a neurodevelopmental disorder. Microglia, the immune cells in central nervous system, related to many neurodevelopmental diseases. Microglia could affect neuronal survival, neuronal death and synaptic plasticity during neurodevelopment. Anomalous microglia during neurodevelopment may be associated with schizophrenia. Therefore, a hypothesis proposes that the abnormal function of microglia leads to the occurrence of schizophrenia. Nowadays, accumulating experiments between microglia and schizophrenia could afford unparalleled probability to assess this hypothesis. Herein, this review summarizes the latest supporting evidence in order to shed light on the mystery of microglia in schizophrenia.
Collapse
Affiliation(s)
- Jingjing Li
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan International Joint Laboratory of Biological Psychiatry, Zhengzhou, China.,Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| | - Yu Wang
- College of First Clinical, Chongqing Medical University, Chongqing, China
| | - Xiuxia Yuan
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan International Joint Laboratory of Biological Psychiatry, Zhengzhou, China.,Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| | - Yulin Kang
- Institute of Environmental Information, Chinese Research Academy of Environmental Sciences, Beijing, China
| | - Xueqin Song
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan International Joint Laboratory of Biological Psychiatry, Zhengzhou, China.,Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
25
|
McEwan F, Glazier JD, Hager R. The impact of maternal immune activation on embryonic brain development. Front Neurosci 2023; 17:1146710. [PMID: 36950133 PMCID: PMC10025352 DOI: 10.3389/fnins.2023.1146710] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 02/15/2023] [Indexed: 03/08/2023] Open
Abstract
The adult brain is a complex structure with distinct functional sub-regions, which are generated from an initial pool of neural epithelial cells within the embryo. This transition requires a number of highly coordinated processes, including neurogenesis, i.e., the generation of neurons, and neuronal migration. These take place during a critical period of development, during which the brain is particularly susceptible to environmental insults. Neurogenesis defects have been associated with the pathogenesis of neurodevelopmental disorders (NDDs), such as autism spectrum disorder and schizophrenia. However, these disorders have highly complex multifactorial etiologies, and hence the underlying mechanisms leading to aberrant neurogenesis continue to be the focus of a significant research effort and have yet to be established. Evidence from epidemiological studies suggests that exposure to maternal infection in utero is a critical risk factor for NDDs. To establish the biological mechanisms linking maternal immune activation (MIA) and altered neurodevelopment, animal models have been developed that allow experimental manipulation and investigation of different developmental stages of brain development following exposure to MIA. Here, we review the changes to embryonic brain development focusing on neurogenesis, neuronal migration and cortical lamination, following MIA. Across published studies, we found evidence for an acute proliferation defect in the embryonic MIA brain, which, in most cases, is linked to an acceleration in neurogenesis, demonstrated by an increased proportion of neurogenic to proliferative divisions. This is accompanied by disrupted cortical lamination, particularly in the density of deep layer neurons, which may be a consequence of the premature neurogenic shift. Although many aspects of the underlying pathways remain unclear, an altered epigenome and mitochondrial dysfunction are likely mechanisms underpinning disrupted neurogenesis in the MIA model. Further research is necessary to delineate the causative pathways responsible for the variation in neurogenesis phenotype following MIA, which are likely due to differences in timing of MIA induction as well as sex-dependent variation. This will help to better understand the underlying pathogenesis of NDDs, and establish therapeutic targets.
Collapse
|
26
|
Garrido-Torres N, Cerrillos L, García Cerro S, Pérez Gómez A, Canal-Rivero M, de Felipe B, Alameda L, Marqués Rodríguez R, Anillo S, Praena J, Duque Sánchez C, Roca C, Paniagua M, López Díaz A, Romero-García R, Olbrich P, Puertas Albarracín MDP, Reguera Pozuelo P, Sosa IL, Moreno Dueñas MB, Pineda Cachero R, Zamudio Juan L, García Rumi V, Guerrero Benitez M, Figueroa R, Martín Rendón AM, Partida A, Rodríguez Cocho MI, Gallardo Trujillo C, Gallego Jiménez I, García Spencer S, Gómez Verdugo M, Bermejo Fernández C, Pérez Benito M, Castillo Reina RE, Cejudo López A, Sánchez Tomás C, Chacón Gamero MÁ, Rubio A, Moreno Mellado A, Ramos Herrero V, Starr E, González Fernández de Palacios M, García Victori E, Pavón Delgado A, Fernández Cuervo I, Arias Ruiz A, Menéndez Gil IE, Domínguez Gómez I, Coca Mendoza I, Ayesa-Arriola R, Fañanas L, Leza JC, Cisneros JM, Sánchez Céspedes J, Ruiz-Mateos E, Crespo-Facorro B, Ruiz-Veguilla M. Examining the immune signatures of SARS-CoV-2 infection in pregnancy and the impact on neurodevelopment: Protocol of the SIGNATURE longitudinal study. Front Pediatr 2022; 10:899445. [PMID: 36619503 PMCID: PMC9811261 DOI: 10.3389/fped.2022.899445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 10/26/2022] [Indexed: 12/24/2022] Open
Abstract
The COVID-19 pandemic represents a valuable opportunity to carry out cohort studies that allow us to advance our knowledge on pathophysiological mechanisms of neuropsychiatric diseases. One of these opportunities is the study of the relationships between inflammation, brain development and an increased risk of suffering neuropsychiatric disorders. Based on the hypothesis that neuroinflammation during early stages of life is associated with neurodevelopmental disorders and confers a greater risk of developing neuropsychiatric disorders, we propose a cohort study of SARS-CoV-2-infected pregnant women and their newborns. The main objective of SIGNATURE project is to explore how the presence of prenatal SARS-CoV-2 infection and other non-infectious stressors generates an abnormal inflammatory activity in the newborn. The cohort of women during the COVID-19 pandemic will be psychological and biological monitored during their pregnancy, delivery, childbirth and postpartum. The biological information of the umbilical cord (foetus blood) and peripheral blood from the mother will be obtained after childbirth. These samples and the clinical characterisation of the cohort of mothers and newborns, are tremendously valuable at this time. This is a protocol report and no analyses have been conducted yet, being currently at, our study is in the recruitment process step. At the time of this publication, we have identified 1,060 SARS-CoV-2 infected mothers and all have already given birth. From the total of identified mothers, we have recruited 537 SARS-COV-2 infected women and all of them have completed the mental health assessment during pregnancy. We have collected biological samples from 119 mothers and babies. Additionally, we have recruited 390 non-infected pregnant women.
Collapse
Affiliation(s)
- Nathalia Garrido-Torres
- Mental Health Unit, Virgen del Rocio University Hospital, Seville, Spain
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
- Spanish Network for Research in Mental Health CIBERSAM, ISCIII, Madrid, Spain
- Department of Psychiatry, University of Seville, Seville, Spain
| | - Lucas Cerrillos
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Susana García Cerro
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
- Spanish Network for Research in Mental Health CIBERSAM, ISCIII, Madrid, Spain
| | - Alberto Pérez Gómez
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Seville Biomedical Research Institute (IBiS), Virgen del Rocío University Hospital, CSIC, University of Seville, Seville, Spain
| | - Manuel Canal-Rivero
- Mental Health Unit, Virgen del Rocio University Hospital, Seville, Spain
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
- Spanish Network for Research in Mental Health CIBERSAM, ISCIII, Madrid, Spain
- Department of Psychiatry, University of Seville, Seville, Spain
| | - Beatriz de Felipe
- Congenital Immunity Disorders Group de Alteraciones Congénitas de Inmunidad, Seville Biomedical Research Institute, Seville, Spain
- Pediatrics, Infectious Diseases and Immunology Department, University Hospital Virgen del Rocío, Sevilla, Spain
| | - Luis Alameda
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
- Department of Psychiatry, University of Seville, Seville, Spain
- Service of General Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Renata Marqués Rodríguez
- Department of Pediatrics, Virgen del Rocío University Hospital / Institute of Biomedicine of Seville (IBiS), Seville, Spain
| | - Sergio Anillo
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Julia Praena
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Cristina Duque Sánchez
- Department of Pediatrics, Virgen del Rocío University Hospital / Institute of Biomedicine of Seville (IBiS), Seville, Spain
| | - Cristina Roca
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Seville Biomedical Research Institute (IBiS), Virgen del Rocío University Hospital, CSIC, University of Seville, Seville, Spain
| | - María Paniagua
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Seville Biomedical Research Institute (IBiS), Virgen del Rocío University Hospital, CSIC, University of Seville, Seville, Spain
| | - Alvaro López Díaz
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
- Spanish Network for Research in Mental Health CIBERSAM, ISCIII, Madrid, Spain
- Department of Psychiatry, University of Seville, Seville, Spain
| | - Rafael Romero-García
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
- Spanish Network for Research in Mental Health CIBERSAM, ISCIII, Madrid, Spain
- Department of Medical Physiology and Biophysics, Seville Biomedical Research Institute (IBiS), Virgen del Rocío University Hospital, CSIC, University of Seville, Seville, Spain
| | - Peter Olbrich
- Congenital Immunity Disorders Group de Alteraciones Congénitas de Inmunidad, Seville Biomedical Research Institute, Seville, Spain
| | | | - Pablo Reguera Pozuelo
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
| | - Irene Luján Sosa
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - María Begoña Moreno Dueñas
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Rocío Pineda Cachero
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Lidia Zamudio Juan
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Verónica García Rumi
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Mercedes Guerrero Benitez
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Rosario Figueroa
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Antonio Manuel Martín Rendón
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Antonio Partida
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - María Isabel Rodríguez Cocho
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Carmen Gallardo Trujillo
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Isabel Gallego Jiménez
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Sarah García Spencer
- Department of Pediatrics, Virgen del Rocío University Hospital / Institute of Biomedicine of Seville (IBiS), Seville, Spain
| | - Marta Gómez Verdugo
- Department of Pediatrics, Virgen del Rocío University Hospital / Institute of Biomedicine of Seville (IBiS), Seville, Spain
| | - Cintia Bermejo Fernández
- Department of Pediatrics, Virgen del Rocío University Hospital / Institute of Biomedicine of Seville (IBiS), Seville, Spain
| | - María Pérez Benito
- Department of Pediatrics, Virgen del Rocío University Hospital / Institute of Biomedicine of Seville (IBiS), Seville, Spain
| | | | - Angela Cejudo López
- Department of family medicine, Virgen del Rocío University Hospital, Primary Care Health Centers, Seville, Spain
| | - Candela Sánchez Tomás
- Department of family medicine, Virgen del Rocío University Hospital, Primary Care Health Centers, Seville, Spain
| | | | - Ana Rubio
- Mental Health Unit, Virgen del Rocio University Hospital, Seville, Spain
| | - Amanda Moreno Mellado
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
| | - Víctor Ramos Herrero
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
| | - Ella Starr
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
| | | | - Elena García Victori
- Department of Pediatrics, Virgen del Rocío University Hospital / Institute of Biomedicine of Seville (IBiS), Seville, Spain
| | - Antonio Pavón Delgado
- Department of Pediatrics, Virgen del Rocío University Hospital / Institute of Biomedicine of Seville (IBiS), Seville, Spain
| | | | | | | | | | | | - Rosa Ayesa-Arriola
- Spanish Network for Research in Mental Health CIBERSAM, ISCIII, Madrid, Spain
- Department of Psychiatry, University Hospital Marqués de Valdecilla - Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Lourdes Fañanas
- Spanish Network for Research in Mental Health CIBERSAM, ISCIII, Madrid, Spain
- Department of Evolutionary Biology, Ecology and Environmental Sciences, Faculty of Biology, University of Barcelona (UB), Barcelona, Spain
| | - Juan C Leza
- Spanish Network for Research in Mental Health CIBERSAM, ISCIII, Madrid, Spain
- Department of Pharmacology & Toxicology, Faculty of Medicine, Universidad Complutense Madrid, CIBERSAM, Imas12, IUINQ, Madrid, Spain
| | - José M Cisneros
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Seville Biomedical Research Institute (IBiS), Virgen del Rocío University Hospital, CSIC, University of Seville, Seville, Spain
| | - Javier Sánchez Céspedes
- Viral Diseases and Infections in Immunodeficiencies Research Group, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Seville, Spain
| | - Ezequiel Ruiz-Mateos
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Seville Biomedical Research Institute (IBiS), Virgen del Rocío University Hospital, CSIC, University of Seville, Seville, Spain
| | - Benedicto Crespo-Facorro
- Mental Health Unit, Virgen del Rocio University Hospital, Seville, Spain
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
- Spanish Network for Research in Mental Health CIBERSAM, ISCIII, Madrid, Spain
- Department of Psychiatry, University of Seville, Seville, Spain
| | - Miguel Ruiz-Veguilla
- Mental Health Unit, Virgen del Rocio University Hospital, Seville, Spain
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
- Spanish Network for Research in Mental Health CIBERSAM, ISCIII, Madrid, Spain
- Department of Psychiatry, University of Seville, Seville, Spain
| |
Collapse
|
27
|
Rodrigues-Neves AC, Ambrósio AF, Gomes CA. Microglia sequelae: brain signature of innate immunity in schizophrenia. Transl Psychiatry 2022; 12:493. [PMID: 36443303 PMCID: PMC9705537 DOI: 10.1038/s41398-022-02197-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 11/29/2022] Open
Abstract
Schizophrenia is a psychiatric disorder with significant impact on individuals and society. The current pharmacologic treatment, which principally alleviates psychosis, is focused on neurotransmitters modulation, relying on drugs with severe side effects and ineffectiveness in a significant percentage of cases. Therefore, and due to difficulties inherent to diagnosis and treatment, it is vital to reassess alternative cellular and molecular drug targets. Distinct risk factors - genetic, developmental, epigenetic, and environmental - have been associated with disease onset and progression, giving rise to the proposal of different pathophysiological mechanisms and putative pharmacological targets. Immunity is involved and, particularly microglia - innate immune cells of the central nervous system, critically involved in brain development - have captured attention as cellular players. Microglia undergo marked morphologic and functional alterations in the human disease, as well as in animal models of schizophrenia, as reported in several original papers. We cluster the main findings of clinical studies by groups of patients: (1) at ultra-high risk of psychosis, (2) with a first episode of psychosis or recent-onset schizophrenia, and (3) with chronic schizophrenia; in translational studies, we highlight the time window of appearance of particular microglia alterations in the most well studied animal model in the field (maternal immune activation). The organization of clinical and translational findings based on schizophrenia-associated microglia changes in different phases of the disease course may help defining a temporal pattern of microglia changes and may drive the design of novel therapeutic strategies.
Collapse
Affiliation(s)
- A. Catarina Rodrigues-Neves
- grid.8051.c0000 0000 9511 4342Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Univ Coimbra, Faculty of Pharmacy, Coimbra, Portugal
| | - António. F. Ambrósio
- grid.8051.c0000 0000 9511 4342Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Catarina A. Gomes
- grid.8051.c0000 0000 9511 4342Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Univ Coimbra, Faculty of Pharmacy, Coimbra, Portugal
| |
Collapse
|
28
|
Hydrogen Sulfide Attenuates the Cognitive Dysfunction in Parkinson's Disease Rats via Promoting Hippocampal Microglia M2 Polarization by Enhancement of Hippocampal Warburg Effect. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2792348. [PMID: 35028004 PMCID: PMC8752224 DOI: 10.1155/2022/2792348] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/06/2021] [Accepted: 11/15/2021] [Indexed: 01/08/2023]
Abstract
Identification of innovative therapeutic targets for the treatment of cognitive impairment in Parkinson's disease (PD) is urgently needed. Hydrogen sulfide (H2S) plays an important role in cognitive function. Therefore, this work is aimed at investigating whether H2S attenuates the cognitive impairment in PD and the underlying mechanisms. In the rotenone- (ROT-) established PD rat model, NaHS (a donor of H2S) attenuated the cognitive impairment and promoted microglia polarization from M1 towards M2 in the hippocampus of PD rats. NaHS also dramatically upregulated the Warburg effect in the hippocampus of PD rats. 2-Deoxyglucose (2-DG, an inhibitor of the Warburg effect) abolished NaHS-upregulated Warburg effect in the hippocampus of PD rats. Moreover, the inhibited hippocampal Warburg effect by 2-DG abrogated H2S-excited the enhancement of hippocampal microglia M2 polarization and the improvement of cognitive function in ROT-exposed rats. Our data demonstrated that H2S inhibits the cognitive dysfunction in PD via promoting microglia M2 polarization by enhancement of hippocampal Warburg effect.
Collapse
|
29
|
Present and future antipsychotic drugs: a systematic review of the putative mechanisms of action for efficacy and a critical appraisal under a translational perspective. Pharmacol Res 2022; 176:106078. [PMID: 35026403 DOI: 10.1016/j.phrs.2022.106078] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/23/2021] [Accepted: 01/07/2022] [Indexed: 01/10/2023]
Abstract
Antipsychotics represent the mainstay of schizophrenia pharmacological therapy, and their role has been expanded in the last years to mood disorders treatment. Although introduced in 1952, many years of research were required before an accurate picture of how antipsychotics work began to emerge. Despite the well-recognized characterization of antipsychotics in typical and atypical based on their liability to induce motor adverse events, their main action at dopamine D2R to elicit the "anti-psychotic" effect, as well as the multimodal action at other classes of receptors, their effects on intracellular mechanisms starting with receptor occupancy is still not completely understood. Significant lines of evidence converge on the impact of these compounds on multiple molecular signaling pathways implicated in the regulation of early genes and growth factors, dendritic spine shape, brain inflammation, and immune response, tuning overall the function and architecture of the synapse. Here we present, based on PRISMA approach, a comprehensive and systematic review of the above mechanisms under a translational perspective to disentangle those intracellular actions and signaling that may underline clinically relevant effects and represent potential targets for further innovative strategies in antipsychotic therapy.
Collapse
|
30
|
Bao M, Hofsink N, Plösch T. LPS vs. Poly I:C Model: Comparison of Long-Term Effects of Bacterial and Viral Maternal Immune Activation (MIA) on the Offspring. Am J Physiol Regul Integr Comp Physiol 2021; 322:R99-R111. [PMID: 34874190 PMCID: PMC8782664 DOI: 10.1152/ajpregu.00087.2021] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A prominent health issue nowadays is the COVID-19 pandemic, which poses acute risks to human health. However, the long-term health consequences are largely unknown and cannot be neglected. An especially vulnerable period for infection is pregnancy, when infections could have long-term health effect on the child. Evidence suggests that maternal immune activation (MIA) induced by either bacteria or viruses presents various effects on the offspring, leading to adverse phenotypes in many organ systems. This review compares the mechanisms of bacterial and viral MIA and the possible long-term outcomes for the offspring by summarizing the outcome in animal LPS and Poly I:C models. Both models are activated immune responses mediated by Toll-like receptors. The outcomes for MIA offspring include neurodevelopment, immune response, circulation, metabolism, and reproduction. Some of these changes continue to exist until later life. Besides different doses and batches of LPS and Poly I:C, the injection day, administration route, and also different animal species influence the outcomes. Here, we specifically aim to support colleagues when choosing their animal models for future studies.
Collapse
Affiliation(s)
- Mian Bao
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Naomi Hofsink
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Torsten Plösch
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Perinatal Neurobiology, Department of Human Medicine, School of Medicine and Health Sciences Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
31
|
Talukdar PM, Abdul F, Maes M, Berk M, Venkatasubramanian G, Kutty BM, Debnath M. A proof-of-concept study of maternal immune activation mediated induction of Toll-like receptor (TLR) and inflammasome pathways leading to neuroprogressive changes and schizophrenia-like behaviours in offspring. Eur Neuropsychopharmacol 2021; 52:48-61. [PMID: 34261013 DOI: 10.1016/j.euroneuro.2021.06.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/17/2021] [Accepted: 06/21/2021] [Indexed: 11/18/2022]
Abstract
Infection, particularly prenatal infection, leads to an enhanced risk of schizophrenia in the offspring. Interestingly, few data exist on the pathway(s) such as TLR and inflammasome, primarily involved in sensing the microorganisms and inducing downstream inflammatory responses, apoptosis and neuroprogressive changes that drive prenatal infection-induced risk of schizophrenia. Herein, we aimed to discern whether prenatal infection-induced maternal immune activation (MIA) causes schizophrenia-like behaviours through activation of TLR and inflammasome pathways in the brain of offspring. Sprague Dawley rats (n=15/group) were injected either with poly (I:C) or LPS or saline at gestational day (GD)-12. Significantly elevated plasma levels of IL-6, TNF-α and IL-17A assessed after 24 hours were observed in both the poly (I:C) and LPS-treated rats, while IL-1β was only elevated in LPS-treated rats, indicating MIA. The offspring of poly (I:C)-and LPS-treated dams displayed increased anxiety-like behaviours, deficits in social behaviours and prepulse inhibition. The hippocampus of offspring rats showed increased expression of Tlr3, Tlr4, Nlrp3, Il1b, and Il18 of poly (I:C) and Tlr4, Nlrp3, Cas1, Il1b, and Il18 of LPS-treated dams. Furthermore, Tlr and inflammasome genes were associated with social deficits and impaired prepulse inhibition in offspring rats. The results suggest that MIA due to prenatal infection can trigger TLR and inflammasome pathways and enhances the risk of schizophrenia-like behaviours in the later stages of life of the offspring.
Collapse
Affiliation(s)
- Pinku Mani Talukdar
- Department of Human Genetics, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Fazal Abdul
- Department of Human Genetics, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria; Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Michael Berk
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, The Centre of Excellence in Youth Mental Health, the Department of Psychiatry and the Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Australia
| | - Ganesan Venkatasubramanian
- Department of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Bindu M Kutty
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Monojit Debnath
- Department of Human Genetics, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India.
| |
Collapse
|
32
|
Romero-Miguel D, Casquero-Veiga M, MacDowell KS, Torres-Sanchez S, Garcia-Partida JA, Lamanna-Rama N, Romero-Miranda A, Berrocoso E, Leza JC, Desco M, Soto-Montenegro ML. A Characterization of the Effects of Minocycline Treatment During Adolescence on Structural, Metabolic, and Oxidative Stress Parameters in a Maternal Immune Stimulation Model of Neurodevelopmental Brain Disorders. Int J Neuropsychopharmacol 2021; 24:734-748. [PMID: 34165516 PMCID: PMC8453277 DOI: 10.1093/ijnp/pyab036] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 06/01/2021] [Accepted: 06/18/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Minocycline (MIN) is a tetracycline with antioxidant, anti-inflammatory, and neuroprotective properties. Given the likely involvement of inflammation and oxidative stress (IOS) in schizophrenia, MIN has been proposed as a potential adjuvant treatment in this pathology. We tested an early therapeutic window, during adolescence, as prevention of the schizophrenia-related deficits in the maternal immune stimulation (MIS) animal model. METHODS On gestational day 15, Poly I:C or vehicle was injected in pregnant Wistar rats. A total 93 male offspring received MIN (30 mg/kg) or saline from postnatal day (PND) 35-49. At PND70, rats were submitted to the prepulse inhibition test. FDG-PET and T2-weighted MRI brain studies were performed at adulthood. IOS markers were evaluated in frozen brain tissue. RESULTS MIN treatment did not prevent prepulse inhibition test behavioral deficits in MIS offspring. However, MIN prevented morphometric abnormalities in the third ventricle but not in the hippocampus. Additionally, MIN reduced brain metabolism in cerebellum and increased it in nucleus accumbens. Finally, MIN reduced the expression of iNOS (prefrontal cortex, caudate-putamen) and increased the levels of KEAP1 (prefrontal cortex), HO1 and NQO1 (amygdala, hippocampus), and HO1 (caudate-putamen). CONCLUSIONS MIN treatment during adolescence partially counteracts volumetric abnormalities and IOS deficits in the MIS model, likely via iNOS and Nrf2-ARE pathways, also increasing the expression of cytoprotective enzymes. However, MIN treatment during this peripubertal stage does not prevent sensorimotor gating deficits. Therefore, even though it does not prevent all the MIS-derived abnormalities evaluated, our results suggest the potential utility of early treatment with MIN in other schizophrenia domains.
Collapse
Affiliation(s)
| | - Marta Casquero-Veiga
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain,CIBER de Salud Mental (CIBERSAM), Madrid, Spain
| | - Karina S MacDowell
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense (UCM), IIS Imas12, IUIN, Madrid, Spain
| | - Sonia Torres-Sanchez
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Neuropsychopharmacology and Psychobiology Research Group, Psychobiology Area, Department of Psychology, Universidad de Cádiz, Puerto Real (Cádiz), Spain,Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | - José Antonio Garcia-Partida
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Neuropsychopharmacology and Psychobiology Research Group, Psychobiology Area, Department of Psychology, Universidad de Cádiz, Puerto Real (Cádiz), Spain,Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | | | | | - Esther Berrocoso
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Neuropsychopharmacology and Psychobiology Research Group, Psychobiology Area, Department of Psychology, Universidad de Cádiz, Puerto Real (Cádiz), Spain,Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | - Juan C Leza
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense (UCM), IIS Imas12, IUIN, Madrid, Spain
| | - Manuel Desco
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain,CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Leganés, Spain,Centro Nacional de Investigaciones Cardiovasculares, CNIC, Madrid, Spain,Correspondence: Manuel Desco, PhD, Laboratorio de Imagen Médica, Unidad de Medicina y Cirugía Experimental, Hospital General Universitario Gregorio Marañón, Dr. Esquerdo, 46. E-28007 Madrid, Spain ()
| | - María Luisa Soto-Montenegro
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain,CIBER de Salud Mental (CIBERSAM), Madrid, Spain,High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut), University Rey Juan Carlos (URJC), Alcorcón, Spain
| |
Collapse
|
33
|
MacDowell KS, Munarriz-Cuezva E, Meana JJ, Leza JC, Ortega JE. Paliperidone Reversion of Maternal Immune Activation-Induced Changes on Brain Serotonin and Kynurenine Pathways. Front Pharmacol 2021; 12:682602. [PMID: 34054556 PMCID: PMC8156415 DOI: 10.3389/fphar.2021.682602] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 04/30/2021] [Indexed: 12/23/2022] Open
Abstract
Emerging evidence indicates that early-life exposure to environmental factors may increase the risk for schizophrenia via inflammatory mechanisms. Inflammation can alter the metabolism of tryptophan through the oxidative kynurenine pathway to compounds with neurotoxic and neuroprotective activity and compromise serotonin (5-HT) synthesis. Here we investigate the role of serotonergic and kynurenine pathways in the maternal immune activation (MIA) animal model of schizophrenia. The potential reversion exerted by long-term antipsychotic treatment was also evaluated. MIA was induced by prenatal administration of polyinosinic:polycytidylic acid (poly (I:C)) in mice. Expression of different proteins and the content of different metabolites involved in the function of serotonergic and kynurenine pathways was assessed by RT-PCR, immunoblot and ELISA analyses in frontal cortex of the offspring after puberty. MIA decreased tissue 5-HT content and promoted changes in the expression of serotonin transporter, 5-HT2A and 5-HT2C receptors. Expression of indoleamine 2,3-dioxygenase 2 (IDO2) and kynurenine 3-monooxygenase (KMO) was increased by poly (I:C) whereas kynurenine aminotransferase II and its metabolite kynurenic acid were not altered. Long-term paliperidone was able to counteract MIA-induced changes in 5-HT and KMO, and to increase tryptophan availability and tryptophan hydroxylase-2 expression in poly (I:C) mice but not in controls. MIA-induced increase of the cytotoxic risk ratio of kynurenine metabolites (quinolinic/kynurenic acid) was also reversed by paliperidone. MIA induces specific long-term brain effects on serotonergic activity. Such effects seem to be related with alternative activation of the kynurenine metabolic pathway towards a cytotoxic status. Atypical antipsychotic paliperodine partially remediates abnormalities observed after MIA.
Collapse
Affiliation(s)
- Karina S MacDowell
- Department of Pharmacology and Toxicology, Faculty of Medicine, University Complutense of Madrid (UCM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, Bizkaia, Madrid, Spain.,Instituto de Investigación Hospital 12 de Octubre (i+12), IUIN-UCM, Madrid, Spain
| | - Eva Munarriz-Cuezva
- Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, Bizkaia, Madrid, Spain.,Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, Spain
| | - J Javier Meana
- Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, Bizkaia, Madrid, Spain.,Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, Spain.,Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain
| | - Juan C Leza
- Department of Pharmacology and Toxicology, Faculty of Medicine, University Complutense of Madrid (UCM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, Bizkaia, Madrid, Spain.,Instituto de Investigación Hospital 12 de Octubre (i+12), IUIN-UCM, Madrid, Spain
| | - Jorge E Ortega
- Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, Bizkaia, Madrid, Spain.,Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, Spain.,Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain
| |
Collapse
|
34
|
Ketharanathan T, Pereira A, Reets U, Walker D, Sundram S. Brain changes in NF-κB1 and epidermal growth factor system markers at peri-pubescence in the spiny mouse following maternal immune activation. Psychiatry Res 2021; 295:113564. [PMID: 33229121 DOI: 10.1016/j.psychres.2020.113564] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 11/05/2020] [Indexed: 10/23/2022]
Abstract
Environmental risk factors that operate at foetal or neonatal levels increase the vulnerability to schizophrenia, plausibly via stress-immune activation that perturbs the epidermal growth factor (EGF) system, a system critical for neurodevelopment. We investigated potential associations between environmental insults and immune and EGF system changes through a maternal immune activation (MIA) model, using the precocial spiny mice (Acomys cahirinus). After mid-gestation MIA prepubescent offspring showed elevated NF-κB1 protein in nucleus accumbens, decreased EGFR in caudate putamen and a trend for increased PI3K-110δ in ventral hippocampus. Thus, prenatal stress may cause a heightened NF-κB1-mediated immune attenuation of EGF system signalling.
Collapse
Affiliation(s)
- Tharini Ketharanathan
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia.
| | - Avril Pereira
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia; Department of Psychiatry, University of Melbourne, Parkville, VIC 3052, Australia
| | - Udani Reets
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3168, Australia
| | - David Walker
- School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC 3083, Australia
| | - Suresh Sundram
- Department of Psychiatry, Monash University, Clayton, VIC 3168, Australia
| |
Collapse
|
35
|
Tendilla-Beltrán H, Sanchez-Islas NDC, Marina-Ramos M, Leza JC, Flores G. The prefrontal cortex as a target for atypical antipsychotics in schizophrenia, lessons of neurodevelopmental animal models. Prog Neurobiol 2020; 199:101967. [PMID: 33271238 DOI: 10.1016/j.pneurobio.2020.101967] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 10/10/2020] [Accepted: 11/22/2020] [Indexed: 02/06/2023]
Abstract
Prefrontal cortex (PFC) inflammatory imbalance, oxidative/nitrosative stress (O/NS) and impaired neuroplasticity in schizophrenia are thought to have neurodevelopmental origins. Animal models are not only useful to test this hypothesis, they are also effective to establish a relationship among brain disturbances and behavior with the atypical antipsychotics (AAPs) effects. Here we review data of PFC post-mortem and in vivo neuroimaging, human induced pluripotent stem cells (hiPSC), and peripheral blood studies of inflammatory, O/NS, and neuroplasticity alterations in the disease as well as about their modulation by AAPs. Moreover, we reviewed the PFC alterations and the AAP mechanisms beyond their canonical antipsychotic action in four neurodevelopmental animal models relevant to the study of schizophrenia with a distinct approach in the generation of schizophrenia-like phenotypes, but all converge in O/NS and altered neuroplasticity in the PFC. These animal models not only reinforce the neurodevelopmental risk factor model of schizophrenia but also arouse some novel potential therapeutic targets for the disease including the reestablishment of the antioxidant response by the perineuronal nets (PNNs) and the nuclear factor erythroid 2-related factor (Nrf2) pathway, as well as the dendritic spine dynamics in the PFC pyramidal cells.
Collapse
Affiliation(s)
- Hiram Tendilla-Beltrán
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico; Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), CDMX, Mexico
| | | | - Mauricio Marina-Ramos
- Departamento de Ciencias de la Salud, Universidad Popular Autónoma del Estado de Puebla, Puebla, Mexico
| | - Juan C Leza
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Instituto Universitario de Investigación en Neuroquímica (IUIN), UCM. Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Sanitaria Hospital, 12 de Octubre (Imas12), Madrid, Spain
| | - Gonzalo Flores
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico.
| |
Collapse
|
36
|
Toll-like receptors in Alzheimer's disease. J Neuroimmunol 2020; 348:577362. [DOI: 10.1016/j.jneuroim.2020.577362] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/04/2020] [Accepted: 08/15/2020] [Indexed: 02/07/2023]
|
37
|
Zemba Cilic A, Zemba M, Cilic M, Balenovic I, Strbe S, Ilic S, Vukojevic J, Zoricic Z, Filipcic I, Kokot A, Drmic D, Blagaic AB, Tvrdeic A, Seiwerth S, Sikiric P. Pentadecapeptide BPC 157 counteracts L-NAME-induced catalepsy. BPC 157, L-NAME, L-arginine, NO-relation, in the suited rat acute and chronic models resembling 'positive-like' symptoms of schizophrenia. Behav Brain Res 2020; 396:112919. [PMID: 32956773 DOI: 10.1016/j.bbr.2020.112919] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 08/15/2020] [Accepted: 09/14/2020] [Indexed: 12/13/2022]
Abstract
In the suited rat-models, we focused on the stable pentadecapeptide BPC 157, L-NAME, NOS-inhibitor, and L-arginine, NOS-substrate, relation, the effect on schizophrenia-like symptoms. Medication (mg/kg intraperitoneally) was L-NAME (5), L-arginine (100), BPC 157 (0.01), given alone and/or together, at 5 min before the challenge for the acutely disturbed motor activity (dopamine-indirect/direct agonists (amphetamine (3.0), apomorphine (2.5)), NMDA-receptor non-competitive antagonist (MK-801 (0.2)), or catalepsy, (dopamine-receptor antagonist haloperidol (2.0)). Alternatively, BPC 157 10 μg/kg was given immediately after L-NAME 40 mg/kg intraperitoneally. To induce or prevent sensitization, we used chronic methamphetamine administration, alternating 3 days during the first 3 weeks, and challenge after next 4 weeks, and described medication (L-NAME, L-arginine, BPC 157) at 5 min before the methamphetamine at the second and third week. Given alone, BPC 157 or L-arginine counteracted the amphetamine-, apomorphine-, and MK-801-induced effect, haloperidol-induced catalepsy and chronic methamphetamine-induced sensitization. L-NAME did not affect the apomorphine-, and MK-801-induced effects, haloperidol-induced catalepsy and chronic methamphetamine-induced sensitization, but counteracted the acute amphetamine-induced effect. In combinations (L-NAME + L-arginine), as NO-specific counteraction, L-NAME counteracts L-arginine-induced counteractions in the apomorphine-, MK-801-, haloperidol- and methamphetamine-rats, but not in amphetamine-rats. Unlike L-arginine, BPC 157 maintains its counteracting effect in the presence of the NOS-blockade (L-NAME + BPC 157) or NO-system-over-stimulation (L-arginine + BPC 157). Illustrating the BPC 157-L-arginine relationships, BPC 157 restored the antagonization (L-NAME + L-arginine + BPC 157) when it had been abolished by the co-administration of L-NAME with L-arginine (L-NAME + L-arginine). Finally, BPC 157 directly inhibits the L-NAME high dose-induced catalepsy. Further studies would determine precise BPC 157/dopamine/glutamate/NO-system relationships and clinical application.
Collapse
Affiliation(s)
- Andrea Zemba Cilic
- Departments of Pharmacology and Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Mladen Zemba
- Departments of Pharmacology and Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Matija Cilic
- Departments of Pharmacology and Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Igor Balenovic
- Departments of Pharmacology and Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Sanja Strbe
- Departments of Pharmacology and Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Spomenko Ilic
- Departments of Pharmacology and Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Jaksa Vukojevic
- Departments of Pharmacology and Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Zoran Zoricic
- Departments of Pharmacology and Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Igor Filipcic
- Departments of Pharmacology and Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Antonio Kokot
- Departments of Pharmacology and Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Domagoj Drmic
- Departments of Pharmacology and Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Alenka Boban Blagaic
- Departments of Pharmacology and Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ante Tvrdeic
- Departments of Pharmacology and Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Sven Seiwerth
- Departments of Pharmacology and Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Predrag Sikiric
- Departments of Pharmacology and Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia.
| |
Collapse
|
38
|
Haddad FL, Patel SV, Schmid S. Maternal Immune Activation by Poly I:C as a preclinical Model for Neurodevelopmental Disorders: A focus on Autism and Schizophrenia. Neurosci Biobehav Rev 2020; 113:546-567. [PMID: 32320814 DOI: 10.1016/j.neubiorev.2020.04.012] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 01/28/2020] [Accepted: 04/09/2020] [Indexed: 12/18/2022]
Abstract
Maternal immune activation (MIA) in response to a viral infection during early and mid-gestation has been linked through various epidemiological studies to a higher risk for the child to develop autism or schizophrenia-related symptoms.. This has led to the establishment of the pathogen-free poly I:C-induced MIA animal model for neurodevelopmental disorders, which shows relatively high construct and face validity. Depending on the experimental variables, particularly the timing of poly I:C administration, different behavioural and molecular phenotypes have been described that relate to specific symptoms of neurodevelopmental disorders such as autism spectrum disorder and/or schizophrenia. We here review and summarize epidemiological evidence for the effects of maternal infection and immune activation, as well as major findings in different poly I:C MIA models with a focus on poly I:C exposure timing, behavioural and molecular changes in the offspring, and characteristics of the model that relate it to autism spectrum disorder and schizophrenia.
Collapse
Affiliation(s)
- Faraj L Haddad
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada.
| | - Salonee V Patel
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada.
| | - Susanne Schmid
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada.
| |
Collapse
|
39
|
Oxidation-reduction mechanisms in psychiatric disorders: A novel target for pharmacological intervention. Pharmacol Ther 2020; 210:107520. [PMID: 32165136 DOI: 10.1016/j.pharmthera.2020.107520] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 03/02/2020] [Indexed: 12/16/2022]
Abstract
While neurotransmitter dysfunction represents a key component in mental illnesses, there is now a wide agreement for a central pathophysiological hub that includes hormones, neuroinflammation, redox mechanisms as well as oxidative stress. With respect to oxidation-reduction (redox) mechanisms, preclinical and clinical evidence suggests that an imbalance in the pro/anti-oxidative homeostasis toward the increased production of substances with oxidizing potential may contribute to the etiology and manifestation of different psychiatric disorders. The substantial and continous demand for energy renders the brain highly susceptible to disturbances in its energy supply, especially following exposure to stressful events, which may lead to overproduction of reactive oxygen and nitrogen species under conditions of perturbed antioxidant defenses. This will eventually induce different molecular alterations, including extensive protein and lipid peroxidation, increased blood-brain barrier permeability and neuroinflammation, which may contribute to the changes in brain function and morphology observed in mental illnesses. This view may also reconcile different key concepts for psychiatric disorders, such as the neurodevelopmental origin of these diseases, as well as the vulnerability of selective cellular populations that are critical for specific functional abnormalities. The possibility to pharmacologically modulate the redox system is receiving increasing interest as a novel therapeutic strategy to counteract the detrimental effects of the unbalance in brain oxidative mechanisms. This review will describe the main mechanisms and mediators of the redox system and will examine the alterations of oxidative stress found in animal models of psychiatric disorders as well as in patients suffering from mental illnesses, such as schizophrenia and major depressive disorder. In addition, it will discuss studies that examined the effects of psychotropic drugs, including antipsychotics and antidepressants, on the oxidative balance as well as studies that investigated the effectiveness of a direct modulation of oxidative mechanisms in counteracting the behavioral and functional alterations associated with psychiatric disorders, which supports the promising role of the redox system as a novel therapeutic target for the improved treatment of brain disorders.
Collapse
|
40
|
Gogos A, Sbisa A, Witkamp D, van den Buuse M. Sex differences in the effect of maternal immune activation on cognitive and psychosis-like behaviour in Long Evans rats. Eur J Neurosci 2020; 52:2614-2626. [PMID: 31901174 DOI: 10.1111/ejn.14671] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 12/16/2019] [Accepted: 12/23/2019] [Indexed: 02/06/2023]
Abstract
Maternal immune activation during pregnancy is associated with increased risk of development of schizophrenia in later life. There are sex differences in schizophrenia, particularly in terms of age of onset, course of illness and severity of symptoms. However, there is limited and inconsistent literature on sex differences in the effects of maternal immune activation on behaviour with relevance to schizophrenia. The aim of this study was therefore to investigate sex differences in the effects of maternal immune activation by treating Long Evans rats with poly(I:C) on gestational day 15. We compared adult male and female offspring on spatial working memory in the touchscreen trial-unique nonmatching-to-location task, pairwise discrimination and reversal learning, as well as on prepulse inhibition and psychotropic drug-induced locomotor hyperactivity. Male, but not female poly(I:C) offspring displayed a deficit in spatial working memory, particularly at the longer delay. Neither pairwise discrimination nor reversal learning showed an effect of poly(I:C), but female controls outperformed male controls in the reversal learning task. Significant reduction of prepulse inhibition and enhancement of acute methamphetamine-induced locomotor hyperactivity was found similarly in male and female poly(I:C) offspring. These results show that maternal immune activation induces a range of behavioural effects in the offspring, with sex specificity in the effects of maternal immune activation on some aspects of cognition, but not psychosis-like behaviour.
Collapse
Affiliation(s)
- Andrea Gogos
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Vic., Australia
| | - Alyssa Sbisa
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Vic., Australia.,School of Psychology and Public Health, La Trobe University, Melbourne, Vic., Australia
| | - Diede Witkamp
- School of Psychology and Public Health, La Trobe University, Melbourne, Vic., Australia
| | - Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Melbourne, Vic., Australia.,Department of Pharmacology, University of Melbourne, Melbourne, Vic., Australia.,The College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia
| |
Collapse
|
41
|
Aguilar-Valles A, Rodrigue B, Matta-Camacho E. Maternal Immune Activation and the Development of Dopaminergic Neurotransmission of the Offspring: Relevance for Schizophrenia and Other Psychoses. Front Psychiatry 2020; 11:852. [PMID: 33061910 PMCID: PMC7475700 DOI: 10.3389/fpsyt.2020.00852] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/04/2020] [Indexed: 12/21/2022] Open
Abstract
Prenatal infections have been linked to the development of schizophrenia (SCZ) and other neurodevelopmental disorders in the offspring, and work in animal models indicates that this is to occur through the maternal inflammatory response triggered by infection. Several studies in animal models demonstrated that acute inflammatory episodes are sufficient to trigger brain alterations in the adult offspring, especially in the mesolimbic dopamine (DA) system, involved in the pathophysiology of SCZ and other disorders involving psychosis. In the current review, we synthesize the literature on the clinical studies implicating prenatal infectious events in the development of SCZ. Then, we summarize evidence from animal models of maternal immune activation (MIA) and the behavioral and molecular alterations relevant for the function of the DAergic system. Furthermore, we discuss the evidence supporting the involvement of maternal cytokines, such as interleukin 6 (IL-6) and leptin (a hormone with effects on inflammation) in mediating the effects of MIA on the fetal brain, leading to the long-lasting effects on the offspring. In particular, IL-6 has been involved in mediating the effects of MIA animal models in the offspring through actions on the placenta, induction of IL-17a, or triggering the decrease in non-heme iron (hypoferremia). Maternal infection is very likely interacting with additional genetic and environmental risk factors in the development of SCZ; systematically investigating how these interactions produce specific phenotypes is the next step in understanding the etiology of complex psychiatric disorders.
Collapse
Affiliation(s)
| | - Brandon Rodrigue
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | | |
Collapse
|
42
|
Wesołowska A, Jastrzębska-Więsek M, Cios A, Partyka A. The preclinical discovery and development of paliperidone for the treatment of schizophrenia. Expert Opin Drug Discov 2019; 15:279-292. [DOI: 10.1080/17460441.2020.1682994] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Anna Wesołowska
- Jagiellonian University Medical College, Department of Clinical Pharmacy, Kraków, Poland
| | | | - Agnieszka Cios
- Jagiellonian University Medical College, Department of Clinical Pharmacy, Kraków, Poland
| | - Anna Partyka
- Jagiellonian University Medical College, Department of Clinical Pharmacy, Kraków, Poland
| |
Collapse
|
43
|
Robertson OD, Coronado NG, Sethi R, Berk M, Dodd S. Putative neuroprotective pharmacotherapies to target the staged progression of mental illness. Early Interv Psychiatry 2019; 13:1032-1049. [PMID: 30690898 DOI: 10.1111/eip.12775] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 12/26/2018] [Indexed: 12/22/2022]
Abstract
AIM Neuropsychiatric disorders including depression, bipolar and schizophrenia frequently exhibit a neuroprogressive course from prodrome to chronicity. There are a range of agents exhibiting capacity to attenuate biological mechanisms associated with neuroprogression. This review will update the evidence for putative neuroprotective agents including clinical efficacy, mechanisms of action and limitations in current assessment tools, and identify novel agents with neuroprotective potential. METHOD Data for this review were sourced from online databases PUBMED, Embase and Web of Science. Only data published since 2012 were included in this review, no data were excluded based on language or publication origin. RESULTS Each of the agents reviewed inhibit one or multiple pathways of neuroprogression including: inflammatory gene expression and cytokine release, oxidative and nitrosative stress, mitochondrial dysfunction, neurotrophin dysregulation and apoptotic signalling. Some demonstrate clinical efficacy in preventing neural damage or loss, relapse or cognitive/functional decline. Agents include: the psychotropic medications lithium, second generation antipsychotics and antidepressants; other pharmacological agents such as minocycline, aspirin, cyclooxygenase-2 inhibitors, statins, ketamine and alpha-2-delta ligands; and others such as erythropoietin, oestrogen, leptin, N-acetylcysteine, curcumin, melatonin and ebselen. CONCLUSIONS Signals of evidence of clinical neuroprotection are evident for a number of candidate agents. Adjunctive use of multiple agents may present a viable avenue to clinical realization of neuroprotection. Definitive prospective studies of neuroprotection with multimodal assessment tools are required.
Collapse
Affiliation(s)
- Oliver D Robertson
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Victoria, Australia.,Mental Health, Drugs and Alcohol Services, University Hospital Geelong, Barwon Health, Geelong, Victoria, Australia
| | - Nieves G Coronado
- Unidad de Gestión Clinica Salud Mental, Hospital Universitario Virgen del Rocio, Sevilla, Spain
| | - Rickinder Sethi
- Department of Psychiatry, Western University, London, Ontario, Canada
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Victoria, Australia.,Mental Health, Drugs and Alcohol Services, University Hospital Geelong, Barwon Health, Geelong, Victoria, Australia.,Department of Psychiatry, The University of Melbourne, Parkville, Victoria, Australia.,Mood Disorders Research Program, Orygen, the National Centre of Excellence in Youth Mental Health, Parkville, Victoria, Australia.,Department of Psychiatry, Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Seetal Dodd
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Victoria, Australia.,Mental Health, Drugs and Alcohol Services, University Hospital Geelong, Barwon Health, Geelong, Victoria, Australia.,Department of Psychiatry, The University of Melbourne, Parkville, Victoria, Australia.,Mood Disorders Research Program, Orygen, the National Centre of Excellence in Youth Mental Health, Parkville, Victoria, Australia
| |
Collapse
|
44
|
Casquero-Veiga M, García-García D, MacDowell KS, Pérez-Caballero L, Torres-Sánchez S, Fraguas D, Berrocoso E, Leza JC, Arango C, Desco M, Soto-Montenegro ML. Risperidone administered during adolescence induced metabolic, anatomical and inflammatory/oxidative changes in adult brain: A PET and MRI study in the maternal immune stimulation animal model. Eur Neuropsychopharmacol 2019; 29:880-896. [PMID: 31229322 DOI: 10.1016/j.euroneuro.2019.05.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 04/30/2019] [Accepted: 05/29/2019] [Indexed: 12/22/2022]
Abstract
Inflammation and oxidative stress (IOS) are considered key pathophysiological elements in the development of mental disorders. Recent studies demonstrated that the antipsychotic risperidone elicits an antiinflammatory effect in the brain. We administered risperidone for 2-weeks at adolescence to assess its role in preventing brain-related IOS changes in the maternal immune stimulation (MIS) model at adulthood. We also investigated the development of volumetric and neurotrophic abnormalities in areas related to the HPA-axis. Poly I:C (MIS) or saline (Sal) were injected into pregnant Wistar rats on GD15. Male offspring received risperidone or vehicle daily from PND35-PND49. We studied 4 groups (8-15 animals/group): Sal-vehicle, MIS-vehicle, Sal-risperidone and MIS-risperidone. [18F]FDG-PET and MRI studies were performed at adulthood and analyzed using SPM12 software. IOS and neurotrophic markers were measured using WB and ELISA assays in brain tissue. Risperidone elicited a protective function of schizophrenia-related IOS deficits. In particular, risperidone elicited the following effects: reduced volume in the ventricles and the pituitary gland; reduced glucose metabolism in the cerebellum, periaqueductal gray matter, and parietal cortex; higher FDG uptake in the cingulate cortex, hippocampus, thalamus, and brainstem; reduced NFκB activity and iNOS expression; and increased enzymatic activity of CAT and SOD in some brain areas. Our study suggests that some schizophrenia-related IOS changes can be prevented in the MIS model. It also stresses the need to search for novel strategies based on anti-inflammatory compounds in risk populations at early stages in order to alter the course of the disease.
Collapse
Affiliation(s)
- Marta Casquero-Veiga
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; CIBER de Salud Mental (CIBERSAM), Madrid, Spain
| | - David García-García
- Department of Bioengineering and Aerospace Engineering, Universidad Carlos III de Madrid, Leganés, Spain; Facultad de Ciencia y Tecnología, Universidad Isabel I, Burgos, Spain
| | - Karina S MacDowell
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Department of Pharmacology & Toxicology, School of Medicine, Universidad Complutense (UCM), IIS Imas12, IUIN, Madrid, Spain
| | - Laura Pérez-Caballero
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Neuropsychopharmacology & Psychobiology Research Group, Psychobiology Area, Department of Psychology, Universidad de Cádiz, Puerto Real (Cádiz), Spain; Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | - Sonia Torres-Sánchez
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain; Neuropsychopharmacology & Psychobiology Research Group, Universidad de Cádiz, Cádiz, Spain
| | - David Fraguas
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Department of Child and Adolescent Psychiatry, Hospital General Universitario Gregorio Marañón, School of Medicine, Universidad Complutense (UCM), Madrid, Spain
| | - Esther Berrocoso
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Neuropsychopharmacology & Psychobiology Research Group, Psychobiology Area, Department of Psychology, Universidad de Cádiz, Puerto Real (Cádiz), Spain; Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | - Juan C Leza
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Department of Pharmacology & Toxicology, School of Medicine, Universidad Complutense (UCM), IIS Imas12, IUIN, Madrid, Spain
| | - Celso Arango
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Department of Child and Adolescent Psychiatry, Hospital General Universitario Gregorio Marañón, School of Medicine, Universidad Complutense (UCM), Madrid, Spain
| | - Manuel Desco
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Department of Bioengineering and Aerospace Engineering, Universidad Carlos III de Madrid, Leganés, Spain; CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares, CNIC, Madrid, Spain.
| | - María Luisa Soto-Montenegro
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; CIBER de Salud Mental (CIBERSAM), Madrid, Spain
| |
Collapse
|
45
|
Microglial Activation and Psychotic Disorders: Evidence from Pre-clinical and Clinical Studies. Curr Top Behav Neurosci 2019; 44:161-205. [PMID: 30828767 DOI: 10.1007/7854_2018_81] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Clinical and pre-clinical studies have demonstrated an important role of neuroinflammation in the etiology of schizophrenia. While the underlying mechanisms remain poorly understood, there are some studies demonstrating an association between maternal immune activation and behavioral changes in adult offspring and identifying early life infection as a trigger for schizophrenia; in addition, inflammatory markers were found to be increased in the schizophrenic post-mortem brain. During maternal immune activation, pro-inflammatory mediators such as cytokines, chemokines, antibodies, and acute-phase proteins are released in the maternal bloodstream, thus increasing the permeability of the placental barrier and the fetal blood-brain barrier, allowing the inflammatory mediators to enter the fetal brain. In the central nervous system (CNS), these pro-inflammatory mediators are able to activate microglial cells that can release pro-inflammatory cytokines such as tumor necrosis factor alpha (TNF-α), interleukin (IL)-1β, and IL-6. As a consequence, circulating immune cells may infiltrate the brain, increasing cytokine levels and releasing antibodies that aggravate the neuroinflammation. Neuroinflammation may affect processes that are pivotal for normal brain maturation such as myelination, synaptic pruning, and neuronal remodeling. Microglial cell activation and pro-inflammatory mediators have been extensively studied in schizophrenic post-mortem brain samples. Some results of these investigations demonstrated an increase in microglial activation markers, cytokines, and chemokines in post-mortem brain samples from individuals with schizophrenia. In contrast, there are studies that have demonstrated low levels of microglial activation makers in the schizophrenic post-mortem brain. Thus, based on the important role of neuroinflammation as a trigger in the development of schizophrenia, this chapter aims (1) to enumerate evidence of neuroinflammation and microglial activation from pre-clinical schizophrenia models, (2) to show links between schizophrenia and neuroinflammation in clinical studies, and (3) to identify mechanisms by which microglial activation may influence in the development of schizophrenia.
Collapse
|
46
|
García S, Alberich S, MacDowell KS, Martínez-Cengotitabengoa M, López P, Zorrilla I, Leza JC, González-Pinto A. Association Between Medication Adherence and Oxidative Stress in Patients With First-Episode Mania. Front Psychiatry 2019; 10:162. [PMID: 30971964 PMCID: PMC6445053 DOI: 10.3389/fpsyt.2019.00162] [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: 11/06/2018] [Accepted: 03/05/2019] [Indexed: 11/13/2022] Open
Abstract
Poor adherence is a major problem in patients with manic episodes that impairs functionality and has unknown effects on oxidative stress. The objective of this study was to analyze the relationship between adherence to medication, severity of symptoms and oxidative stress in a sample of patients with a first episode of mania. A longitudinal, 6-month study was performed in 60 patients, who were classified as adherent and non-adherent to medication (mainly antipsychotics). Blood levels of oxidative stress parameters and expression of the antioxidant nuclear transcription factor NRF2 in mononuclear cells of peripheral blood were assessed at baseline and at the end of follow-up. In addition, clinical symptoms and functioning were evaluated. Linear multivariate regression was used to determine the relationship between adherence, oxidative stress, and clinical symptoms. Finally, 44 patients completed follow-up. The results of this study showed that at 6-month follow-up, adherence was significantly associated with better functioning and reduced clinical symptoms. Additionally, more severe symptoms were associated with increased levels of oxidative stress and antioxidant parameters. At study completion, non-adherents exhibited greater levels of antioxidants than adherent patients. In conclusion, poor adherence to medication is associated with a poorer prognosis in the medium term and causes increased antioxidant response.
Collapse
Affiliation(s)
- Saínza García
- Department of Psychiatry, University Hospital of Alava-Santiago, Vitoria, Spain.,Centre for Biomedical Research Network on Mental Health (CIBERSAM), Madrid, Spain.,School of Medicine, University of the Basque Country, Vitoria, Spain
| | - Susana Alberich
- Department of Psychiatry, University Hospital of Alava-Santiago, Vitoria, Spain.,Centre for Biomedical Research Network on Mental Health (CIBERSAM), Madrid, Spain
| | - Karina S MacDowell
- Centre for Biomedical Research Network on Mental Health (CIBERSAM), Madrid, Spain.,Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid, IUIN and IIS Hospital 12 de Octubre, Madrid, Spain
| | - Mónica Martínez-Cengotitabengoa
- Department of Psychiatry, University Hospital of Alava-Santiago, Vitoria, Spain.,Centre for Biomedical Research Network on Mental Health (CIBERSAM), Madrid, Spain.,School of Medicine, University of the Basque Country, Vitoria, Spain.,Psychobiology Department, National Distance Education University (UNED), Vitoria, Spain
| | - Purificación López
- Department of Psychiatry, University Hospital of Alava-Santiago, Vitoria, Spain.,Centre for Biomedical Research Network on Mental Health (CIBERSAM), Madrid, Spain.,School of Medicine, University of the Basque Country, Vitoria, Spain
| | - Iñaki Zorrilla
- Department of Psychiatry, University Hospital of Alava-Santiago, Vitoria, Spain.,Centre for Biomedical Research Network on Mental Health (CIBERSAM), Madrid, Spain.,School of Medicine, University of the Basque Country, Vitoria, Spain
| | - Juan Carlos Leza
- Centre for Biomedical Research Network on Mental Health (CIBERSAM), Madrid, Spain.,Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid, IUIN and IIS Hospital 12 de Octubre, Madrid, Spain
| | - Ana González-Pinto
- Department of Psychiatry, University Hospital of Alava-Santiago, Vitoria, Spain.,Centre for Biomedical Research Network on Mental Health (CIBERSAM), Madrid, Spain.,School of Medicine, University of the Basque Country, Vitoria, Spain
| |
Collapse
|
47
|
Bergdolt L, Dunaevsky A. Brain changes in a maternal immune activation model of neurodevelopmental brain disorders. Prog Neurobiol 2018; 175:1-19. [PMID: 30590095 DOI: 10.1016/j.pneurobio.2018.12.002] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 12/13/2018] [Accepted: 12/20/2018] [Indexed: 12/11/2022]
Abstract
The developing brain is sensitive to a variety of insults. Epidemiological studies have identified prenatal exposure to infection as a risk factor for a range of neurological disorders, including autism spectrum disorder and schizophrenia. Animal models corroborate this association and have been used to probe the contribution of gene-environment interactions to the etiology of neurodevelopmental disorders. Here we review the behavior and brain phenotypes that have been characterized in MIA offspring, including the studies that have looked at the interaction between maternal immune activation and genetic risk factors for autism spectrum disorder or schizophrenia. These phenotypes include behaviors relevant to autism, schizophrenia, and other neurological disorders, alterations in brain anatomy, and structural and functional neuronal impairments. The link between maternal infection and these phenotypic changes is not fully understood, but there is increasing evidence that maternal immune activation induces prolonged immune alterations in the offspring's brain which could underlie epigenetic alterations which in turn may mediate the behavior and brain changes. These concepts will be discussed followed by a summary of the pharmacological interventions that have been tested in the maternal immune activation model.
Collapse
Affiliation(s)
- Lara Bergdolt
- University of Nebraska Medical Center, Neurological Sciences, 985960 Nebraska Medical Center, 68105, Omaha, NE, United States
| | - Anna Dunaevsky
- University of Nebraska Medical Center, Neurological Sciences, 985960 Nebraska Medical Center, 68105, Omaha, NE, United States.
| |
Collapse
|
48
|
MacDowell KS, Pinacho R, Leza JC, Costa J, Ramos B, García-Bueno B. Differential regulation of the TLR4 signalling pathway in post-mortem prefrontal cortex and cerebellum in chronic schizophrenia: Relationship with SP transcription factors. Prog Neuropsychopharmacol Biol Psychiatry 2017; 79:481-492. [PMID: 28803924 DOI: 10.1016/j.pnpbp.2017.08.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 07/28/2017] [Accepted: 08/06/2017] [Indexed: 12/21/2022]
Abstract
Alterations in innate immunity may underlie the pathophysiology of schizophrenia (SZ). Toll-like receptor-4 (TLR4) is a master element of innate immunity. The specificity proteins (SPs), transcription factors recently implicated in SZ, are putative regulatory agents of this. This work was aimed at describing alterations in the TLR4 signalling pathway in postmortem brain prefrontal cortex (PFC) and cerebellum (CB) of 16 chronic SZ patients and 14 controls. The possible association of TLR4 pathway with SP1 and SP4 and SZ negative symptomatology is explored. In PFC, TLR4/myeloid differentiation factor 88 (MyD88)/inhibitory subunit of nuclear factor kappa B alpha (IκBα) protein levels were lower in SZ patients, while nuclear transcription factor-κB (NFκB) activity, cyclooxygenase-2 (COX-2) expression and the lipid peroxidation index malondialdehyde (MDA) appeared increased. The pattern of changes in CB is opposite, except for COX-2 expression that remained augmented and MDA levels unaltered. Network interaction analysis showed that TLR4/MyD88/IκBα/NFκB/COX-2 pathway was coupled in PFC and uncoupled in CB. SP4 co-expressed with TLR4 and NFκB in PFC and both SP1 and SP4 co-expressed with NFκB in CB. In PFC, correlation analysis found an inverse relationship between NFκB and negative symptoms. In summary, we found brain region-specific alterations in the TLR4 signalling pathway in chronic SZ, in which SP transcription factors could participate at different levels. Further studies are required to elucidate the regulatory mechanisms of innate immunity in SZ and its relationship with symptoms.
Collapse
Affiliation(s)
- Karina S MacDowell
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain; Dept. of Pharmacology, Faculty of Medicine, Hospital 12 de Octubre Imas12, IUINQ, University Complutense, 28040 Madrid, Spain
| | - Raquel Pinacho
- Psiquiatria Molecular, Institut de Recerca Sant Joan de Déu, Santa Rosa 39-57, 08950 Esplugues de Llobregat, Spain
| | - Juan C Leza
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain; Dept. of Pharmacology, Faculty of Medicine, Hospital 12 de Octubre Imas12, IUINQ, University Complutense, 28040 Madrid, Spain
| | - Joan Costa
- Banc de Teixits Neurologics, Parc Sanitari Sant Joan de Déu, Sant Boi de Llobregat, 08830 Barcelona, Spain; Parc Sanitari Sant Joan de Déu, Dr. Antoni Pujadas, 42, 08830 Sant Boi de Llobregat, Spain
| | - Belén Ramos
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain; Psiquiatria Molecular, Institut de Recerca Sant Joan de Déu, Santa Rosa 39-57, 08950 Esplugues de Llobregat, Spain; Parc Sanitari Sant Joan de Déu, Dr. Antoni Pujadas, 42, 08830 Sant Boi de Llobregat, Spain; Dept. de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain.
| | - Borja García-Bueno
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain; Dept. of Pharmacology, Faculty of Medicine, Hospital 12 de Octubre Imas12, IUINQ, University Complutense, 28040 Madrid, Spain.
| |
Collapse
|
49
|
Bridging Autism Spectrum Disorders and Schizophrenia through inflammation and biomarkers - pre-clinical and clinical investigations. J Neuroinflammation 2017; 14:179. [PMID: 28870209 PMCID: PMC5584030 DOI: 10.1186/s12974-017-0938-y] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 08/08/2017] [Indexed: 12/15/2022] Open
Abstract
In recent years, evidence supporting a link between inflammation and neuropsychiatric disorders has been mounting. Autism spectrum disorders (ASD) and schizophrenia share some clinical similarities which we hypothesize might reflect the same biological basis, namely, in terms of inflammation. However, the diagnosis of ASD and schizophrenia relies solely on clinical symptoms, and to date, there is no clinically useful biomarker to diagnose or monitor the course of such illnesses. The focus of this review is the central role that inflammation plays in ASD and schizophrenia. It spans from pre-clinical animal models to clinical research and excludes in vitro studies. Four major areas are covered: (1) microglia, the inflammatory brain resident myeloid cells, (2) biomarkers, including circulating cytokines, oxidative stress markers, and microRNA players, known to influence cellular processes at brain and immune levels, (3) effect of anti-psychotics on biomarkers and other predictors of response, and (4) impact of gender on response to immune activation, biomarkers, and response to anti-psychotic treatments.
Collapse
|