1
|
Nani JV, Muotri AR, Hayashi MAF. Peering into the mind: unraveling schizophrenia's secrets using models. Mol Psychiatry 2025; 30:659-678. [PMID: 39245692 DOI: 10.1038/s41380-024-02728-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/10/2024]
Abstract
Schizophrenia (SCZ) is a complex mental disorder characterized by a range of symptoms, including positive and negative symptoms, as well as cognitive impairments. Despite the extensive research, the underlying neurobiology of SCZ remain elusive. To overcome this challenge, the use of diverse laboratory modeling techniques, encompassing cellular and animal models, and innovative approaches like induced pluripotent stem cell (iPSC)-derived neuronal cultures or brain organoids and genetically engineered animal models, has been crucial. Immortalized cellular models provide controlled environments for investigating the molecular and neurochemical pathways involved in neuronal function, while iPSCs and brain organoids, derived from patient-specific sources, offer significant advantage in translational research by facilitating direct comparisons of cellular phenotypes between patient-derived neurons and healthy-control neurons. Animal models can recapitulate the different psychopathological aspects that should be modeled, offering valuable insights into the neurobiology of SCZ. In addition, invertebrates' models are genetically tractable and offer a powerful approach to dissect the core genetic underpinnings of SCZ, while vertebrate models, especially mammals, with their more complex nervous systems and behavioral repertoire, provide a closer approximation of the human condition to study SCZ-related traits. This narrative review provides a comprehensive overview of the diverse modeling approaches, critically evaluating their strengths and limitations. By synthesizing knowledge from these models, this review offers a valuable source for researchers, clinicians, and stakeholders alike. Integrating findings across these different models may allow us to build a more holistic picture of SCZ pathophysiology, facilitating the exploration of new research avenues and informed decision-making for interventions.
Collapse
Affiliation(s)
- João V Nani
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil.
- National Institute for Translational Medicine (INCT-TM, CNPq/FAPESP/CAPES), Ribeirão Preto, Brazil.
| | - Alysson R Muotri
- Department of Pediatrics and Department of Molecular and Cellular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Mirian A F Hayashi
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil.
- National Institute for Translational Medicine (INCT-TM, CNPq/FAPESP/CAPES), Ribeirão Preto, Brazil.
| |
Collapse
|
2
|
Sun J, Du X, Chen Y. Current Progress on Postoperative Cognitive Dysfunction: An Update. J Integr Neurosci 2024; 23:224. [PMID: 39735960 DOI: 10.31083/j.jin2312224] [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: 06/12/2024] [Revised: 07/14/2024] [Accepted: 08/14/2024] [Indexed: 12/31/2024] Open
Abstract
Postoperative cognitive dysfunction (POCD) represents a significant clinical concern, particularly among elderly surgical patients. It is characterized by a decline in cognitive performance, affecting memory, attention, coordination, orientation, verbal fluency, and executive function. This decline in cognitive abilities leads to longer hospital stays and increased mortality. This review provides a comprehensive overview of the current progress in understanding the relevant pathogenic factors, possible pathogenic mechanisms, diagnosing, prevention and treatment of POCD, as well as suggesting future research directions. It discusses neuronal damage, susceptible genes, central cholinergic system, central nervous system (CNS) inflammation, stress response and glucocorticoids, and oxidative stress in the development of POCD, aiming to uncover the pathological mechanism and develop effective treatment strategies for POCD.
Collapse
Affiliation(s)
- Jing Sun
- Department of Anesthesia and Perioperative Medicine, The Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China
| | - Xiaohong Du
- Department of Anesthesia and Perioperative Medicine, The Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China
| | - Yong Chen
- Department of Anesthesia and Perioperative Medicine, The Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China
- Jiangxi Province Key of Laboratory of Anesthesiology, 330006 Nanchang, Jiangxi, China
- Department of Anesthesia and Perioperative Care, University of California San Francisco (UCSF), San Francisco, CA 94158, USA
| |
Collapse
|
3
|
Dastan M, Rajaei Z, Sharifi M, Salehi H. Gallic acid ameliorates LPS-induced memory decline by modulating NF-κB, TNF-α, and Caspase 3 gene expression and attenuating oxidative stress and neuronal loss in the rat hippocampus. Metab Brain Dis 2024; 40:12. [PMID: 39556267 DOI: 10.1007/s11011-024-01441-5] [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: 07/13/2024] [Accepted: 10/19/2024] [Indexed: 11/19/2024]
Abstract
Neuroinflammation and apoptosis play critical roles in the pathogenesis of Alzheimer's disease (AD), which is responsible for most cases of dementia in the elderly people. Gallic acid is a phenolic compound with radical scavenging, anti-inflammatory and anti-apoptotic activities. This study aimed to explore the protective effects of gallic acid on LPS-induced spatial memory impairment and find the underlying mechanisms. Gallic acid was orally administered (100 mg/kg) to male Wistar rats for 12 days. LPS was injected intraperitoneally at a dose of 1 mg/kg on days 8-12. Morris water maze paradigm was used to evaluate spatial learning and memory. The mRNA level of nuclear factor kappa B (NF-κB), tumor necrosis factor-α (TNF-α) and Caspase 3, lipid peroxidation and total thiol level was assessed in the rat hippocampus. Neuronal loss and histological changes were also evaluated in the brain. LPS treatment resulted in spatial learning and memory impairment, upregulation of NF-κB, TNF-α, and Caspase 3 mRNA expression, increased lipid peroxidation, decreased total thiol level, and neuronal loss in the hippocampus. Moreover, treatment with gallic acid at a dosage of 100 mg/kg ameliorated memory decline, reduced the mRNA level of NF-κB, TNF-α, and Caspase 3, decreased lipid peroxidation and increased total thiol level in the hippocampus. Gallic acid also prevented LPS-induced neuronal loss and histological changes in the brain. Conclusively, our study demonstrated that gallic acid exerts neuroprotective effect against LPS-induced memory decline in rats. This outcome could be due to anti-inflammatory, antioxidant, and anti-apoptotic activities of gallic acid.
Collapse
Affiliation(s)
- Maryam Dastan
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ziba Rajaei
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mohammadreza Sharifi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Salehi
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
4
|
Bhat K, Helmholz H, Willumeit-Römer R. Application of an in vitro neuroinflammation model to evaluate the efficacy of magnesium-lithium alloys. Front Cell Neurosci 2024; 18:1485427. [PMID: 39539342 PMCID: PMC11558531 DOI: 10.3389/fncel.2024.1485427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Mg-Li alloys can be promising candidates as bioresorbable Li-releasing implants for bipolar disorder and other neurodegenerative disorders. In order to compare the therapeutic efficacy of conventional Li salts and Li delivered through Mg-Li alloy extracts, we tested an in vitro model based on the neuroinflammation hypothesis of mood disorders (peripheral inflammation inducing neuroinflammation) wherein, a coculture of microglia and astrocytes was treated with conditioned medium from pro-inflammatory macrophages. Two alloys, Mg-1.6Li and Mg-9.5Li, were tested in the form of material extracts and well-known outcomes of Li treatment such as GSK3β phosphorylation (indirect flow cytometry) and influence on inflammation-related gene expression (qPCR) were compared against Li salts. This is the first study demonstrating that Li can increase the phosphorylation of GSK3β in glial cells in the presence of excess Mg. Furthermore, Mg-Li alloys were more effective than Li salts in downregulating IL6 and upregulating the neurotrophin GDNF. Mg had no antagonistic effects toward Li-driven downregulation of astrogliosis markers. Overall, the results provide evidence to support further studies employing Mg-Li alloys for neurological applications.
Collapse
|
5
|
Sæther LS, Ueland T, Haatveit B, Vaskinn A, Bärthel Flaaten C, Mohn C, E.G. Ormerod MB, Aukrust P, Melle I, Steen NE, Andreassen OA, Ueland T. Longitudinal course of inflammatory-cognitive subgroups across first treatment severe mental illness and healthy controls. Psychol Med 2024; 54:1-11. [PMID: 39354711 PMCID: PMC11496234 DOI: 10.1017/s003329172400206x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/17/2024] [Accepted: 06/28/2024] [Indexed: 10/03/2024]
Abstract
BACKGROUND While inflammation is associated with cognitive impairment in severe mental illnesses (SMI), there is substantial heterogeneity and evidence of transdiagnostic subgroups across schizophrenia (SZ) and bipolar (BD) spectrum disorders. There is however, limited knowledge about the longitudinal course of this relationship. METHODS Systemic inflammation (C-Reactive Protein, CRP) and cognition (nine cognitive domains) was measured from baseline to 1 year follow-up in first treatment SZ and BD (n = 221), and healthy controls (HC, n = 220). Linear mixed models were used to evaluate longitudinal changes separately in CRP and cognitive domains specific to diagnostic status (SZ, BD, HC). Hierarchical clustering was applied on the entire sample to investigate the longitudinal course of transdiagnostic inflammatory-cognitive subgroups. RESULTS There were no case-control differences or change in CRP from baseline to follow-up. We confirm previous observations of case-control differences in cognition at both time-points and domain specific stability/improvement over time regardless of diagnostic status. We identified transdiagnostic inflammatory-cognitive subgroups at baseline with differing demographics and clinical severity. Despite improvement in cognition, symptoms and functioning, the higher inflammation - lower cognition subgroup (75% SZ; 48% BD; 38% HC) had sustained inflammation and lower cognition, more symptoms, and lower functioning (SMI only) at follow-up. This was in comparison to a lower inflammation - higher cognition subgroup (25% SZ, 52% BD, 62% HC), where SMI participants showed cognitive functioning at HC level with a positive clinical course. CONCLUSIONS Our findings support heterogenous and transdiagnostic inflammatory-cognitive subgroups that are stable over time, and may benefit from targeted interventions.
Collapse
Affiliation(s)
- Linn Sofie Sæther
- Section for Clinical Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Faculty of Medicine, University of Oslo, Norway
- Thrombosis Research Center (TREC), Division of internal medicine, University hospital of North Norway, Tromsø Norway
| | - Beathe Haatveit
- Section for Clinical Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Anja Vaskinn
- Centre for Research and Education in Forensic Psychiatry, Oslo University Hospital, Oslo, Norway
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Camilla Bärthel Flaaten
- Section for Clinical Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Christine Mohn
- National Centre for Suicide Research and Prevention, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Monica B. E.G. Ormerod
- Section for Clinical Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Faculty of Medicine, University of Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Ingrid Melle
- Section for Clinical Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo Norway
| | - Nils Eiel Steen
- Section for Clinical Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Ole A. Andreassen
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Section for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Norway
| | - Torill Ueland
- Section for Clinical Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| |
Collapse
|
6
|
Tiwari H, Kumar A, Barik MR, Kaur H, Mahajan S, Shukla MK, Gupta M, Yadav G, Nargotra A. Repositioning the existing drugs for neuroinflammation: a fusion of computational approach and biological validation to counter the Parkinson's disease progression. Mol Divers 2024; 28:2759-2770. [PMID: 37542020 DOI: 10.1007/s11030-023-10708-5] [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: 06/05/2023] [Accepted: 07/28/2023] [Indexed: 08/06/2023]
Abstract
Parkinson's disease is caused by the deficiency of striatal dopamine and the accumulation of aggregated α-synuclein in the substantia nigra pars compacta (SNpc). Neuroinflammation associated with oxidative stress is a key factor contributing to the death of dopaminergic neurons in SNpc and advancement of Parkinson's disease. Two molecular targets, i.e., nuclear factor kappa-light-chain-enhancer (NF-kB) and α-synuclein play a substantial role in neuroinflammation progression. Therefore, the compounds targeting these neuroinflammatory targets hold a great potential to combat Parkinson's disease. Thereby, in this study, molecular docking and Connectivity Map (CMap) based gene expression profiling was utilized to reposition the approved drugs as neuroprotective agents for Parkinson's disease. With in silico screening, two drugs namely theophylline and propylthiouracil were selected for anti-neuroinflammatory activity evaluation in in vivo models of chronic neuroinflammation. The neuroinflammatory effect of the identified compounds was confirmed by quantifying the expression of three important neuroinflammatory mediators, i.e. IL-6, TNF-alpha, and IL-1 beta on brain tissue using ELISA assay. The ELISA experiment demonstrated that both compounds significantly decreased the expression of neuroinflammatory mediators, highlighting the compounds' potential in neuroinflammation management. Furthermore, the drug and disease interaction network of the two identified drugs and diseases (neuroinflammation and Parkinson's disease) suggested that the two drugs might interact with various targets namely adenosine receptors, Poly [ADP-ribose] polymerase-1, myeloperoxidase (MPO) and thyroid peroxidase through multiple pathways associated with neuroinflammation and Parkinson's disease. Computational studies suggest that a particular drug may be effective in managing Parkinson's disease associated with neuroinflammation. However, further research is needed to confirm this in biological experiments.
Collapse
Affiliation(s)
- Harshita Tiwari
- Discovery Informatics, NPMC Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Amit Kumar
- Mutagenicity Laboratory, Pharmacology Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Manas Ranjan Barik
- Discovery Informatics, NPMC Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Harjot Kaur
- Discovery Informatics, NPMC Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Shubham Mahajan
- Discovery Informatics, NPMC Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Monu Kumar Shukla
- PK-PD Toxicology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Monika Gupta
- Discovery Informatics, NPMC Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Govind Yadav
- Mutagenicity Laboratory, Pharmacology Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180001, India
| | - Amit Nargotra
- Discovery Informatics, NPMC Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India.
| |
Collapse
|
7
|
Liu Z, Wang Y, Wang S, Wu J, Jia C, Tan X, Liu X, Huang X, Zhang L. Unraveling the causative connection between urticaria, inflammatory cytokines, and mental disorders: Perspectives from genetic evidence. Skin Res Technol 2024; 30:e13906. [PMID: 39300828 PMCID: PMC11413335 DOI: 10.1111/srt.13906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 07/05/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND The genetic association between urticaria and mental disorders and whether inflammatory cytokines mediate this process remains unclear. MATERIALS AND METHODS A Mendelian randomization (MR) approaches to elucidate the causal relationship between urticaria and mental disorders and to validate the mediation of inflammatory cytokines. Genome-wide association study (GWAS) databases used were obtained from Psychiatric Genomics Cooperation (PGC), GWAS Catalog, and FinnGen Consortium. Our study was conducted using inverse variance weighted (IVW) and Bayesian weighted MR (BWMR) methods for joint analysis. RESULTS The MR results showed that urticaria increased the risk of attention deficit hyperactivity disorder (ADHD) (odds ratio [OR] = $ = $ 1.088, 95% confidence interval [CI]: 1.026-1.154, p = $ = $ 0.0051); cholinergic urticaria increased the risk of bipolar disorder (BD) (OR = $ = $ 1.012, 95% CI: 1.001-1.022, p = $ = $ 0.0274); dermatographic urticaria increased the risk of ADHD (OR = $ = $ 1.057, 95% CI: 1.005-1.112, p = $ = $ 0.0323); idiopathic urticaria increased the risk of schizophrenia (SCZ) (OR = $ = $ 1.057, 95% CI: 1.005-1.112, p = $ = $ 0.0323); other unspecified urticaria increased the risk of ADHD (OR = $ = $ 1.085, 95% CI: 1.023-1.151, p = $ = $ 0.0063). We found that eight inflammatory cytokines were negatively associated with mental disorders and seven inflammatory cytokines were positively associated with mental disorders. Finally, our results suggested that inflammatory cytokines do not act as mediators between urticaria and mental disorders. CONCLUSIONS Our study reveals a causal relationship between urticaria and the increased risk of mental disorders. We suggest that the treatment of urticaria could incorporate psychiatric interventions and mental health assessment of patients.
Collapse
Affiliation(s)
- ZhiRong Liu
- Department of General SurgeryChengdu Second People's HospitalChengduChina
| | | | - ShiHao Wang
- School of Biological Science and TechnologyChengdu Medical CollegeChengduChina
| | - JiaXin Wu
- Graduate SchoolChengdu Medical CollegeChengduChina
| | - Cui Jia
- Development and Regeneration Key Laboratory of Sichuan ProvinceInstitute of NeuroscienceDepartment of Pathology and PathophysiologyChengdu Medical CollegeChengduChina
| | - Xuan Tan
- School of Biological Science and TechnologyChengdu Medical CollegeChengduChina
| | - XinLian Liu
- Development and Regeneration Key Laboratory of Sichuan ProvinceInstitute of NeuroscienceDepartment of Pathology and PathophysiologyChengdu Medical CollegeChengduChina
| | - XinWei Huang
- Shanghai Key Laboratory of Anesthesiology and Brain Functional ModulationClinical Research Center for Anesthesiology and Perioperative MedicineTranslational Research Institute of Brain and Brain‐Like IntelligenceShanghai Fourth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
| | - LuShun Zhang
- Development and Regeneration Key Laboratory of Sichuan ProvinceInstitute of NeuroscienceDepartment of Pathology and PathophysiologyChengdu Medical CollegeChengduChina
| |
Collapse
|
8
|
Deng Y, Li Q, Song J, Guo R, Ma T, Liu Z, Liu Q. Intervention effects of low-molecular-weight chondroitin sulfate from the nasal cartilage of yellow cattle on lipopolysaccharide-induced behavioral disorders: regulation of the microbiome-gut-brain axis. Front Nutr 2024; 11:1371691. [PMID: 38835960 PMCID: PMC11148680 DOI: 10.3389/fnut.2024.1371691] [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: 01/16/2024] [Accepted: 04/24/2024] [Indexed: 06/06/2024] Open
Abstract
Chondroitin sulfate (CS) is a sulfated linear polysaccharide with different functional activities, including antioxidant, anti-inflammatory, lipid-lowering, and immune regulation. As natural sulfated polysaccharides have high molecular weight, high apparent viscosity, low water solubility, complex structure, and high negative charge, they have difficulty binding to receptors within cells across tissue barriers, resulting in low bioavailability and unclear structure-activity relationships. In this study, an H2O2-Vc oxidative degradation system was employed to perform environmentally friendly and controllable degradation of CS extracted from the nasal cartilage of Shaanxi Yellow cattle. Two low-molecular-weight chondroitin sulfates (LMWCSs), CS-1 (14.8 kDa) and CS-2 (50.9 kDa), that exhibit strong in vitro free radical scavenging ability were obtained, and their structures were characterized. Mice intraperitoneally administered lipopolysaccharide (LPS) were used to explore the cognitive intervention effects of LMWCS. Supplementing CS-1 and CS-2 significantly downregulated the levels of the serum inflammatory factors, TNF-α and IL-1β, promoted the expression of GSH in the brain, and inhibited the production of the lipid peroxidation product, malondialdehyde (MDA), ultimately inhibiting LPS-induced cognitive impairment in mice. Surprisingly, compared to the LPS model group, the abundances of Streptococcus, Eisenbergiella, Vampirovibrio, Coprococcus, Enterococcus and Lachnoanaerobaculum were significantly increased in the intestines of mice in the CS-1 and CS-2 group, whereas those of Parabacteroides and Mycoplasma were significantly decreased. Altogether, this study provides a theoretical basis for the comprehensive utilization of agricultural and animal resources and the application of brain nutrition, anti-inflammatory, and LMWCS health products.
Collapse
Affiliation(s)
- Yuxuan Deng
- College of Food Science and Technology, Northwest University, Xi'an, China
| | - Qingyuan Li
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Junxian Song
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Rui Guo
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Tianchen Ma
- College of Food Science and Technology, Northwest University, Xi'an, China
| | - Zhigang Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Qian Liu
- College of Food Science and Technology, Northwest University, Xi'an, China
| |
Collapse
|
9
|
Martos D, Lőrinczi B, Szatmári I, Vécsei L, Tanaka M. The Impact of C-3 Side Chain Modifications on Kynurenic Acid: A Behavioral Analysis of Its Analogs in the Motor Domain. Int J Mol Sci 2024; 25:3394. [PMID: 38542368 PMCID: PMC10970565 DOI: 10.3390/ijms25063394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/09/2024] [Accepted: 03/13/2024] [Indexed: 11/11/2024] Open
Abstract
The central nervous system (CNS) is the final frontier in drug delivery because of the blood-brain barrier (BBB), which poses significant barriers to the access of most drugs to their targets. Kynurenic acid (KYNA), a tryptophan (Trp) metabolite, plays an important role in behavioral functions, and abnormal KYNA levels have been observed in neuropsychiatric conditions. The current challenge lies in delivering KYNA to the CNS owing to its polar side chain. Recently, C-3 side chain-modified KYNA analogs have been shown to cross the BBB; however, it is unclear whether they retain the biological functions of the parent molecule. This study examined the impact of KYNA analogs, specifically, SZR-72, SZR-104, and the newly developed SZRG-21, on behavior. The analogs were administered intracerebroventricularly (i.c.v.), and their effects on the motor domain were compared with those of KYNA. Specifically, open-field (OF) and rotarod (RR) tests were employed to assess motor activity and skills. SZR-104 increased horizontal exploratory activity in the OF test at a dose of 0.04 μmol/4 μL, while SZR-72 decreased vertical activity at doses of 0.04 and 0.1 μmol/4 μL. In the RR test, however, neither KYNA nor its analogs showed any significant differences in motor skills at either dose. Side chain modification affects affective motor performance and exploratory behavior, as the results show for the first time. In this study, we showed that KYNA analogs alter emotional components such as motor-associated curiosity and emotions. Consequently, drug design necessitates the development of precise strategies to traverse the BBB while paying close attention to modifications in their effects on behavior.
Collapse
Affiliation(s)
- Diána Martos
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged, Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, H-6725 Szeged, Hungary;
| | - Bálint Lőrinczi
- Institute of Pharmaceutical Chemistry and HUN-REN–SZTE Stereochemistry Research Group, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary; (B.L.); (I.S.)
| | - István Szatmári
- Institute of Pharmaceutical Chemistry and HUN-REN–SZTE Stereochemistry Research Group, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary; (B.L.); (I.S.)
| | - László Vécsei
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged, Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, H-6725 Szeged, Hungary;
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - Masaru Tanaka
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged, Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, H-6725 Szeged, Hungary;
| |
Collapse
|
10
|
Morimoto K, Watanuki S, Eguchi R, Kitano T, Otsuguro KI. Short-term memory impairment following recovery from systemic inflammation induced by lipopolysaccharide in mice. Front Neurosci 2023; 17:1273039. [PMID: 37920299 PMCID: PMC10618367 DOI: 10.3389/fnins.2023.1273039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 09/27/2023] [Indexed: 11/04/2023] Open
Abstract
The relationship between neuroinflammation and mental disorders has been recognized and investigated for over 30 years. Diseases of systemic or peripheral inflammation, such as sepsis, peritonitis, and infection, are associated with increased risk of mental disorders with neuroinflammation. To elucidate the pathogenesis, systemic administration of lipopolysaccharide (LPS) in mice is often used. LPS-injected mice exhibit behavioral abnormalities with glial activation. However, these studies are unlikely to recapitulate the clinical pathophysiology of human patients, as most studies focus on the acute inflammatory response with systemic symptoms occurring within 24 h of LPS injection. In this study, we focus on the effects of LPS on behavioral abnormalities following recovery from systemic symptoms and investigate the mechanisms of pathogenesis. Several behavioral tests were performed in LPS-injected mice, and to assess neuroinflammation, the time course of the morphological change and expression of inflammatory factors in neurons, astrocytes, and microglia were investigated. At 7 days post-LPS injection, mice exhibited short-term memory impairment accompanied by the suppression of neuronal activity and increases in morphologically immature spines. Glial cells were transiently activated in the hippocampus concomitant with upregulation of the microglial phagocytosis marker CD68 3 days after injection. Here we show that transient glial cell activation in the acute response phase affects neuronal activity and behavior following recovery from systemic symptoms. These findings provide novel insights for studies using the LPS-induced inflammation model and that will contribute to the development of treatments for mental disorders of this etiology.
Collapse
Affiliation(s)
- Kohei Morimoto
- Laboratory of Pharmacology, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Shu Watanuki
- Laboratory of Pharmacology, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Ryota Eguchi
- Laboratory of Pharmacology, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Taisuke Kitano
- Laboratory of Veterinary Biochemistry, School of Veterinary Medicine, Kitasato University, Aomori, Japan
| | - Ken-ichi Otsuguro
- Laboratory of Pharmacology, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
11
|
Liu Z, Chen B, Xiang S, Hu S. Self-immolative nanocapsules precisely regulate depressive neuronal microenvironment for synergistic antidepression therapy. J Nanobiotechnology 2023; 21:274. [PMID: 37592281 PMCID: PMC10433581 DOI: 10.1186/s12951-023-02008-9] [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: 02/28/2023] [Accepted: 07/14/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Pharmacotherapy constitutes the first-line treatment for depression. However, its clinical use is hindered by several limitations, such as time lag, side effects, and narrow therapeutic windows. Nanotechnology can be employed to shorten the onset time by ensuring permeation across the blood brain barrier (BBB) to precisely deliver more therapeutic agents; unfortunately, formidable challenges owing to the intrinsic shortcomings of commercial drugs remain. RESULTS Based on the extraordinary capability of monoamines to regulate the neuronal environment, we engineer a network nanocapsule for delivering serotonin (5-hydroxytryptamine, 5-HT) and catalase (CAT) to the brain parenchyma for synergistic antidepression therapy. The nanoantidepressants are fabricated by the formation of 5-HT polymerization and simultaneous payload CAT, following by surface modifications using human serum albumin and rabies virus glycoprotein. The virus-inspired nanocapsules benefit from the surface-modifying strategies and exhibit pronounced BBB penetration. Once nanocapsules reach the brain parenchyma, the mildly acidic conditions trigger the release of 5-HT from the sacrificial nanocapsule. Releasing 5-HT further positively regulate moods, relieving depressive symptoms. Meanwhile, cargo CAT alleviates neuroinflammation and enhances therapeutic efficacy of 5-HT. CONCLUSION Altogether, the results offer detailed information encouraging the rational designing of nanoantidepressants and highlighting the potential of nanotechnology in mental health disorder therapies.
Collapse
Affiliation(s)
- Ziyao Liu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Biological, Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Bei Chen
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Shijun Xiang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Shuo Hu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Key Laboratory of Biological, Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
12
|
van Engelen MPE, Verfaillie SCJ, Dols A, Oudega ML, Boellaard R, Golla SSV, den Hollander M, Ossenkoppele R, Scheltens P, van Berckel BNM, Pijnenburg YAL, Vijverberg EGB. Altered brain metabolism in frontotemporal dementia and psychiatric disorders: involvement of the anterior cingulate cortex. EJNMMI Res 2023; 13:71. [PMID: 37493827 PMCID: PMC10371967 DOI: 10.1186/s13550-023-01020-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/11/2023] [Indexed: 07/27/2023] Open
Abstract
BACKGROUND Behavioural symptoms and frontotemporal hypometabolism overlap between behavioural variant of frontotemporal dementia (bvFTD) and primary psychiatric disorders (PPD), hampering diagnostic distinction. Voxel-wise comparisons of brain metabolism might identify specific frontotemporal-(hypo)metabolic regions between bvFTD and PPD. We investigated brain metabolism in bvFTD and PPD and its relationship with behavioural symptoms, social cognition, severity of depressive symptoms and cognitive functioning. RESULTS Compared to controls, bvFTD showed decreased metabolism in the dorsal anterior cingulate cortex (dACC) (p < 0.001), orbitofrontal cortex (OFC), temporal pole, dorsolateral prefrontal cortex (dlPFC) and caudate, whereas PPD showed no hypometabolism. Compared to PPD, bvFTD showed decreased metabolism in the dACC (p < 0.001, p < 0.05FWE), insula, Broca's area, caudate, thalamus, OFC and temporal cortex (p < 0.001), whereas PPD showed decreased metabolism in the motor cortex (p < 0.001). Across bvFTD and PPD, decreased metabolism in the temporal cortex (p < 0.001, p < 0.05FWE), dACC and frontal cortex was associated with worse social cognition. Decreased metabolism in the dlPFC was associated with compulsiveness (p < 0.001). Across bvFTD, PPD and controls, decreased metabolism in the PFC and motor cortex was associated with executive dysfunctioning (p < 0.001). CONCLUSIONS Our findings indicate subtle but distinct metabolic patterns in bvFTD and PPD, most strongly in the dACC. The degree of frontotemporal and cingulate hypometabolism was related to impaired social cognition, compulsiveness and executive dysfunctioning. Our findings suggest that the dACC might be an important region to differentiate between bvFTD and PPD but needs further validation.
Collapse
Affiliation(s)
- Marie-Paule E van Engelen
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.
| | - Sander C J Verfaillie
- Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Medical Psychology, Amsterdam Public Health Research Institute, University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- GGZ inGeest Specialized Mental Health Care, Amsterdam, The Netherlands
| | - Annemieke Dols
- Department of Psychiatry, UMC Utrecht Brain Center, University of Utrecht, Utrecht, The Netherlands
| | - Mardien L Oudega
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- GGZ inGeest Specialized Mental Health Care, Amsterdam, The Netherlands
- Department of Psychiatry, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Mood, Anxiety, Psychosis, Sleep & Stress Program, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Sandeep S V Golla
- Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Marijke den Hollander
- Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Rik Ossenkoppele
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- EQT Life Sciences Partners, Amsterdam, The Netherlands
| | - Bart N M van Berckel
- Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Yolande A L Pijnenburg
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Everard G B Vijverberg
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
13
|
da Silva BS, Grevet EH, Silva LCF, Ramos JKN, Rovaris DL, Bau CHD. An overview on neurobiology and therapeutics of attention-deficit/hyperactivity disorder. DISCOVER MENTAL HEALTH 2023; 3:2. [PMID: 37861876 PMCID: PMC10501041 DOI: 10.1007/s44192-022-00030-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/29/2022] [Indexed: 10/21/2023]
Abstract
Attention-Deficit/Hyperactivity Disorder (ADHD) is a prevalent psychiatric condition characterized by developmentally inappropriate symptoms of inattention and/or hyperactivity/impulsivity, which leads to impairments in the social, academic, and professional contexts. ADHD diagnosis relies solely on clinical assessment based on symptom evaluation and is sometimes challenging due to the substantial heterogeneity of the disorder in terms of clinical and pathophysiological aspects. Despite the difficulties imposed by the high complexity of ADHD etiology, the growing body of research and technological advances provide good perspectives for understanding the neurobiology of the disorder. Such knowledge is essential to refining diagnosis and identifying new therapeutic options to optimize treatment outcomes and associated impairments, leading to improvements in all domains of patient care. This review is intended to be an updated outline that addresses the etiological and neurobiological aspects of ADHD and its treatment, considering the impact of the "omics" era on disentangling the multifactorial architecture of ADHD.
Collapse
Affiliation(s)
- Bruna Santos da Silva
- ADHD and Developmental Psychiatry Programs, Hospital de Clínicas de Porto Alegre, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
- Department of Genetics and Graduate Program in Genetics and Molecular Biology, Instituto de Biociências, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas Universidade de Sao Paulo, São Paulo, Brazil
- Laboratory of Physiological Genomics of Mental Health (PhysioGen Lab), Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Eugenio Horacio Grevet
- ADHD and Developmental Psychiatry Programs, Hospital de Clínicas de Porto Alegre, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
- Department of Psychiatry and Graduate Program in Psychiatry and Behavioral Sciences, Faculdade de Medicina, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Luiza Carolina Fagundes Silva
- ADHD and Developmental Psychiatry Programs, Hospital de Clínicas de Porto Alegre, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
- Department of Psychiatry and Graduate Program in Psychiatry and Behavioral Sciences, Faculdade de Medicina, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - João Kleber Neves Ramos
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas Universidade de Sao Paulo, São Paulo, Brazil
- Laboratory of Physiological Genomics of Mental Health (PhysioGen Lab), Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Diego Luiz Rovaris
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas Universidade de Sao Paulo, São Paulo, Brazil
- Laboratory of Physiological Genomics of Mental Health (PhysioGen Lab), Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Claiton Henrique Dotto Bau
- ADHD and Developmental Psychiatry Programs, Hospital de Clínicas de Porto Alegre, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil.
- Department of Genetics and Graduate Program in Genetics and Molecular Biology, Instituto de Biociências, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil.
- Department of Psychiatry and Graduate Program in Psychiatry and Behavioral Sciences, Faculdade de Medicina, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil.
| |
Collapse
|
14
|
Norouzi-Barough L, Asgari Khosroshahi A, Gorji A, Zafari F, Shahverdi Shahraki M, Shirian S. COVID-19-Induced Stroke and the Potential of Using Mesenchymal Stem Cells-Derived Extracellular Vesicles in the Regulation of Neuroinflammation. Cell Mol Neurobiol 2023; 43:37-46. [PMID: 35025001 PMCID: PMC8755896 DOI: 10.1007/s10571-021-01169-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 11/09/2021] [Indexed: 02/07/2023]
Abstract
Ischemic stroke (IS) is a known neurological complication of COVID-19 infection, which is associated with high mortality and disability. Following IS, secondary neuroinflammation that occurs can play both harmful and beneficial roles and lead to further injury or repair of damaged neuronal tissue, respectively. Since inflammation plays a pivotal role in the pathogenesis of COVID-19-induced stroke, targeting neuroinflammation could be an effective strategy for modulating the immune responses following ischemic events. Numerous investigations have indicated that the application of mesenchymal stem cells-derived extracellular vesicles (MSC-EVs) improves functional recovery following stroke, mainly through reducing neuroinflammation as well as promoting neurogenesis and angiogenesis. Therefore, MSC-EVs can be applied for the regulation of SARS-CoV-2-mediated inflammation and the management of COVID-19- related ischemic events. In this study, we have first described the advantages and disadvantages of neuroinflammation in the pathological evolution after IS and summarized the characteristics of neuroinflammation in COVID-19-related stroke. Then, we have discussed the potential benefit of MSC-EVs in the regulation of inflammatory responses after COVID-19-induced ischemic events.
Collapse
Affiliation(s)
- Leyla Norouzi-Barough
- Acquired Immunodeficiency Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Ali Gorji
- Epilepsy Research Center, Department of Neurosurgery, Westfälische Wilhelms-Universitat Münster, Munster, Germany
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Fariba Zafari
- Cellular and Molecular Research Center, Research Institute for Prevention of Non- Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
| | | | - Sadegh Shirian
- Department of Pathology, School of Veterinary Medicine, Shahrekord University, Shahrekord, Iran.
- Shiraz Molecular Pathology Research Center, Dr. Daneshbod Pathol Lab, Shiraz, Iran.
| |
Collapse
|
15
|
Bhalla S, Mehan S. 4-hydroxyisoleucine mediated IGF-1/GLP-1 signalling activation prevents propionic acid-induced autism-like behavioural phenotypes and neurochemical defects in experimental rats. Neuropeptides 2022; 96:102296. [PMID: 36307249 DOI: 10.1016/j.npep.2022.102296] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/16/2022] [Accepted: 10/16/2022] [Indexed: 11/06/2022]
Abstract
Autism is a neuropsychiatric disorder characterized by a neurotransmitter imbalance that impairs neurodevelopment processes. Autism development is marked by communication difficulties, poor socio-emotional health, and cognitive impairment. Insulin-like growth factor-1 (IGF-1) and glucagon-like growth factor-1 (GLP-1) are responsible for regular neuronal growth and homeostasis. Autism progression has been linked to dysregulation of IGF-1/GLP-1 signalling. 4-hydroxyisoleucine (HI), a pharmacologically active amino acid produced from Trigonella foenum graecum, works as an insulin mimic and has neuroprotective properties. The GLP-1 analogue liraglutide (LRG) was employed in our investigation to compare the efficacy of 4-HI in autism prevention. The current study explores the protective effects of 4-HI 50 and 100 mg/kg orally on IGF-1/GLP-1 signalling activation in a PPA-induced experimental model of autism. Propionic acid (PPA) injections to rats by intracerebroventricular (ICV) route for the first 11 days of the experiment resulted in autism-like neurobehavioral, neurochemical, gross morphological, and histopathological abnormalities. In addition, we investigated the dose-dependent neuroprotective effects of 4-HI on the levels of several neurotransmitters and neuroinflammatory cytokines in rat brain homogenate and blood plasma. Neuronal apoptotic and anti-oxidant cellular markers were also studied in blood plasma and brain homogenate samples. Furthermore, the luxol fast blue (LFB) staining results demonstrated significant demyelination in the brains of PPA-induced rats reversed by 4-HI treatment. Rats were assessed for spontaneous locomotor impairments, neuromuscular coordination, stress-like behaviour, learning, and memory to assess neurobehavioral abnormalities. The administration of 4-HI and LRG significantly reversed the behavioural, gross and histological abnormalities in the PPA-treated rat brains. After treatment with 4-HI and LRG, LFB-stained photomicrographs of PPA-treated rats' brains demonstrated the recovery of white matter loss. Our findings indicate that 4-HI protects neurons in rats with autism by enhancing the IGF-1 and GLP-1 protein levels.
Collapse
Affiliation(s)
- Sonalika Bhalla
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India.
| |
Collapse
|
16
|
Nani JV, Almeida PGC, Noto C, Bressan RA, Brietzke E, Hayashi MAF. Unraveiling the correlation among neurodevelopmental and inflammatory biomarkers in patients with chronic schizophrenia. Nord J Psychiatry 2022; 76:559-564. [PMID: 36189960 DOI: 10.1080/08039488.2021.2023217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Nuclear distribution element like-1 (Ndel1) is a cytosolic oligopeptidase, which was suggested as a potential biomarker of aberrant neurodevelopment and early stage of schizophrenia (SCZ). The involvement of Ndel1 in neurite outgrowth, neuronal migration and neurodevelopment was demonstrated. Moreover, Ndel1 cleaves neuropeptides, including the endogenous antipsychotic peptide neurotensin, and lower Ndel1 activity was reported in SCZ patients compared with healthy controls (HCs). Changes in brain-derived neurotrophic factor (BDNF) and inflammatory cytokines levels were also implicated in SCZ. OBJECTIVE This preliminary study aimed to investigate the interactions between these immune and neurodevelopmental/neurotrophic biomarkers, namely BDNF and the recently identified SCZ biomarker Ndel1. RESULTS We observed lower Ndel1 activity and IL-4 levels, and higher BDNF levels, in plasma of SCZ (N = 23) compared with HCs (N = 29). Interestingly, significant correlation between Ndel1 activity and IL-4 levels was observed in SCZ, while no correlation with any other evaluated interleukins (namely IL-2, IL-8, IL-10 and IL-17A) or BDNF levels was noticed. CONCLUSION Although this hypothesis needs to be further explored for a better understanding of the mechanisms by which these altered pathways are associated to each other in SCZ, we suggest that Ndel1 and the inflammatory marker IL-4 are directly correlated.
Collapse
Affiliation(s)
- João V Nani
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), Sao Paulo, Brazil.,National Institute for Translational Medicine (INCT-TM, CNPq), Sao Paulo, Brazil
| | - Priscila G C Almeida
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), Sao Paulo, Brazil
| | - Cristiano Noto
- Department of Psychiatry, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), Sao Paulo, Brazil
| | - Rodrigo A Bressan
- Department of Psychiatry, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), Sao Paulo, Brazil
| | - Elisa Brietzke
- Department of Psychiatry, Queen's University School of Medicine, Kingston, Canada
| | - Mirian A F Hayashi
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), Sao Paulo, Brazil.,National Institute for Translational Medicine (INCT-TM, CNPq), Sao Paulo, Brazil
| |
Collapse
|
17
|
Murdaca G, Paladin F, Casciaro M, Vicario CM, Gangemi S, Martino G. Neuro-Inflammaging and Psychopathological Distress. Biomedicines 2022; 10:2133. [PMID: 36140234 PMCID: PMC9495653 DOI: 10.3390/biomedicines10092133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/24/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Inflammaging is a low degree of chronic and systemic tissue inflammation associated with aging, and is intimately linked to pro-inflammatory mediators. These substances are involved in the pathogenesis of chronic inflammatory diseases and related psychopathological symptoms. When inflammation and aging affect the brain, we use the term neuro-inflammaging. In this review, we focused on the neuro-inflammatory process typical of advanced ages and the related psychopathological symptoms, with particular attention to understanding the immune-pathogenetic mechanisms involved and the potential use of immunomodulatory drugs in the control of clinical psychological signs. Inflammation and CNS were demonstrated being intimately linked in the neuro-inflammatory loop. IL-1, IL-6, TNF-a, COX and PGE are only partially responsible. BBB permeability and the consequent oxidative stress resulting from tissue damage make the rest. Some authors elaborated the "theory of cytokine-induced depression". Inflammation has a crucial role in the onset symptoms of psychopathological diseases as it is capable of altering the metabolism of biogenic monoamines involved in their pathogenesis. In recent years, NSAIDs as an adjunct therapy in the treatment of relevant psychopathological disorders associated with chronic inflammatory conditions demonstrated their efficacy. Additionally, novel molecules have been studied, such as adalimumab, infliximab, and etanercept showing antidepressant and anxiolytic promising results. However, we are only at the beginning of a new era characterized by the use of biological drugs for the treatment of inflammatory and autoimmune diseases, and this paper aims to stimulate future studies in such a direction.
Collapse
Affiliation(s)
- Giuseppe Murdaca
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, 16132 Genoa, Italy
| | - Francesca Paladin
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, 16132 Genoa, Italy
| | - Marco Casciaro
- Department of Biomedical and Dental Science and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy
| | | | - Sebastiano Gangemi
- Allergy and Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Gabriella Martino
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| |
Collapse
|
18
|
Clausen AR, Durand S, Petersen RL, Staunstrup NH, Qvist P. Circulating miRNAs as Potential Biomarkers for Patient Stratification in Bipolar Disorder: A Combined Review and Data Mining Approach. Genes (Basel) 2022; 13:1038. [PMID: 35741801 PMCID: PMC9222282 DOI: 10.3390/genes13061038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/02/2022] [Accepted: 06/07/2022] [Indexed: 02/01/2023] Open
Abstract
Bipolar disorder is a debilitating psychiatric condition that is shaped in a concerted interplay between hereditary and triggering risk factors. Profound depression and mania define the disorder, but high clinical heterogeneity among patients complicates diagnosis as well as pharmacological intervention. Identification of peripheral biomarkers that capture the genomic response to the exposome may thus progress the development of personalized treatment. MicroRNAs (miRNAs) play a prominent role in of post-transcriptional gene regulation in the context of brain development and mental health. They are coordinately modulated by multifarious effectors, and alteration in their expression profile has been reported in a variety of psychiatric conditions. Intriguingly, miRNAs can be released from CNS cells and enter circulatory bio-fluids where they remain remarkably stable. Hence, peripheral circulatory miRNAs may act as bio-indicators for the combination of genetic risk, environmental exposure, and/or treatment response. Here we provide a comprehensive literature search and data mining approach that summarize current experimental evidence supporting the applicability of miRNAs for patient stratification in bipolar disorder.
Collapse
Affiliation(s)
- Alexandra R. Clausen
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (A.R.C.); (S.D.); (R.L.P.); (N.H.S.)
| | - Simon Durand
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (A.R.C.); (S.D.); (R.L.P.); (N.H.S.)
| | - Rasmus L. Petersen
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (A.R.C.); (S.D.); (R.L.P.); (N.H.S.)
| | - Nicklas H. Staunstrup
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (A.R.C.); (S.D.); (R.L.P.); (N.H.S.)
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, 8000 Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, 8000 Aarhus, Denmark
- Centre for Genomics and Personalized Medicine, CGPM, Aarhus University, 8000 Aarhus, Denmark
- Blood Bank and Immunology, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Per Qvist
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (A.R.C.); (S.D.); (R.L.P.); (N.H.S.)
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, 8000 Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, 8000 Aarhus, Denmark
- Centre for Genomics and Personalized Medicine, CGPM, Aarhus University, 8000 Aarhus, Denmark
- Department of Health Science and Technology, Aalborg University, 9200 Aalborg, Denmark
| |
Collapse
|
19
|
Nguyen LTH, Choi MJ, Shin HM, Yang IJ. Coptisine Alleviates Imiquimod-Induced Psoriasis-like Skin Lesions and Anxiety-like Behavior in Mice. Molecules 2022; 27:1412. [PMID: 35209199 PMCID: PMC8878104 DOI: 10.3390/molecules27041412] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 02/04/2023] Open
Abstract
Psoriasis is a common inflammatory skin disorder, which can be associated with psychological disorders, such as anxiety and depression. This study investigated the efficacy and the mechanism of action of a natural compound coptisine using imiquimod (IMQ)-induced psoriasis mice. Coptisine reduced the severity of psoriasis-like skin lesions, decreased epidermal hyperplasia and the levels of inflammatory cytokines TNF-α, IL-17, and IL-22. Furthermore, coptisine improved IMQ-induced anxiety in mice by increasing the number of entries and time in open arms in the elevated plus maze (EPM) test. Coptisine also lowered the levels of inflammatory cytokines TNF-α and IL-1β in the prefrontal cortex of psoriasis mice. HaCaT keratinocytes and BV2 microglial cells were used to investigate the effects of coptisine in vitro. In M5-treated HaCaT cells, coptisine decreased the production of IL-6, MIP-3α/CCL20, IP-10/CXCL10, and ICAM-1 and suppressed the NF-κB signaling pathway. In LPS-stimulated BV2 cells, coptisine reduced the secretion of TNF-α and IL-1β. These findings suggest that coptisine might be a potential candidate for psoriasis treatment by improving both disease severity and psychological comorbidities.
Collapse
Affiliation(s)
| | | | - Heung-Mook Shin
- Department of Physiology, College of Korean Medicine, Dongguk University, Gyeongju 38066, Korea; (L.T.H.N.); (M.-J.C.)
| | - In-Jun Yang
- Department of Physiology, College of Korean Medicine, Dongguk University, Gyeongju 38066, Korea; (L.T.H.N.); (M.-J.C.)
| |
Collapse
|
20
|
Malashenkova I, Ushakov V, Zakharova N, Krynskiy S, Ogurtsov D, Hailov N, Chekulaeva E, Ratushnyy A, Kartashov S, Kostyuk G, Didkovsky N. Neuro-Immune Aspects of Schizophrenia with Severe Negative Symptoms: New Diagnostic Markers of Disease Phenotype. Sovrem Tekhnologii Med 2021; 13:24-33. [PMID: 35265356 PMCID: PMC8858398 DOI: 10.17691/stm2021.13.6.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Indexed: 11/30/2022] Open
Abstract
The aim of the study was to analyze the immune-inflammatory profile of patients with paranoid schizophrenia and relate it to the severity of negative symptoms and the MRI data in order to identify biomarkers of schizophrenia severity, search for new approaches to therapy, and control its effectiveness. Materials and Methods The main group included 51 patients with paranoid schizophrenia, the control group - 30 healthy subjects. Patients underwent MRI scans and immunological studies, which included an assessment of natural and adaptive immunity, the systemic level of key pro-inflammatory and anti-inflammatory cytokines, and other markers of inflammation. Results Disorders of immunity and immunoinflammatory profile in patients with paranoid schizophrenia with severe negative symptoms were revealed for the first time: in the presence of severe negative symptoms (>15 points according to the NSA-4 scale), the levels of humoral immunity factors, cytokines IL-10 and IL-12p40 and neurotrophin NGF were increased as well as the markers of systemic inflammation. Morphometric changes in the brain, typical for patients with schizophrenia, and also specific for patients with severe negative symptoms, were determined. The data analysis revealed correlations between the immune changes with structural changes in some of the brain areas, including the frontal cortex and hippocampus. Associations were found between the levels of anti-inflammatory IL-10, IL-12p40 cytokines and morphometric parameters of the brain, specific only for schizophrenic patients with severe negative symptoms. Conclusion The interdisciplinary approach, combining brain morphometry with in-depth immunological and clinical studies, made it possible to determine neurobiological, immune, and neurocognitive markers of paranoid schizophrenia with severe negative symptoms. The results are important for further deciphering the pathogenesis of schizophrenia and its subtypes, as well as for the search for new approaches to the treatment of severe forms of the disease.
Collapse
Affiliation(s)
- I.K. Malashenkova
- Head of the Laboratory of Molecular Immunology and Virology; National Research Center “Kurchatov Institute”, 1 Akademika Kurchatova Square, Moscow, 123182, Russia; Leading Researcher, Laboratory of Clinical Immunology; Federal Research and Clinical Center of Physical-Chemical Medicine
| | - V.L. Ushakov
- Associate Professor, Senior Researcher; National Research Nuclear University MEPhI, 31 Kashirskoe Shosse, Moscow, 115409, Russia; Department Head; Alekseev Psychiatric Clinical Hospital No.1, Moscow Department of Health, 2 Zagorodnoe Shosse, Moscow, 117152, Russia; Leading Researcher, Institute for Advanced Brain Research; Lomonosov Moscow State University, 27/1 Lomonosov Avenue, Moscow, 119192, Russia
| | - N.V. Zakharova
- Head of the Laboratory for Fundamental Research Methods; Alekseev Psychiatric Clinical Hospital No.1, Moscow Department of Health, 2 Zagorodnoe Shosse, Moscow, 117152, Russia
| | - S.A. Krynskiy
- Researcher, Laboratory of Molecular Immunology and Virology; National Research Center “Kurchatov Institute”, 1 Akademika Kurchatova Square, Moscow, 123182, Russia
| | - D.P. Ogurtsov
- Researcher, Laboratory of Molecular Immunology and Virology; National Research Center “Kurchatov Institute”, 1 Akademika Kurchatova Square, Moscow, 123182, Russia; Researcher, Laboratory of Clinical Immunology; Federal Research and Clinical Center of Physical-Chemical Medicine
| | - N.A. Hailov
- Senior Researcher, Resource Center for Molecular and Cellular Biology; National Research Center “Kurchatov Institute”, 1 Akademika Kurchatova Square, Moscow, 123182, Russia
| | - E.I. Chekulaeva
- Junior Researcher, Resource Center for Molecular and Cellular Biology; National Research Center “Kurchatov Institute”, 1 Akademika Kurchatova Square, Moscow, 123182, Russia
| | - A.Y. Ratushnyy
- Researcher, Laboratory of Cell Physiology; Russian Federation State Research Center Institute of Biomedical Problems of the Russian Academy of Sciences, 76A Khoroshevskoe Shosse, Moscow, 123007, Russia
| | - S.I. Kartashov
- Laboratory Deputy Head; National Research Center “Kurchatov Institute”, 1 Akademika Kurchatova Square, Moscow, 123182, Russia
| | - G.P. Kostyuk
- Professor, Chief Physician; Alekseev Psychiatric Clinical Hospital No.1, Moscow Department of Health, 2 Zagorodnoe Shosse, Moscow, 117152, Russia
| | - N.A. Didkovsky
- Professor, Head of the Laboratory of Clinical Immunology; Federal Research and Clinical Center of Physical-Chemical Medicine
| |
Collapse
|
21
|
Jeon P, Mackinley M, Théberge J, Palaniyappan L. The trajectory of putative astroglial dysfunction in first episode schizophrenia: a longitudinal 7-Tesla MRS study. Sci Rep 2021; 11:22333. [PMID: 34785674 PMCID: PMC8595701 DOI: 10.1038/s41598-021-01773-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 11/03/2021] [Indexed: 11/28/2022] Open
Abstract
Myo-inositol is mainly found in astroglia and its levels has been shown to be reduced in the anterior cingulate cortex (ACC) of patients with schizophrenia. We investigate the status of astroglial integrity indexed by ACC myo-inositol at the onset and over the first 6 months of treatment of first episode schizophrenia. We employed 7 T magnetic resonance spectroscopy (1H-MRS) and quantified myo-inositol spectra at the dorsal ACC in 31 participants; 21 patients with schizophrenia with median lifetime antipsychotic exposure of less than 3 days, followed up after 6 months of treatment, and 10 healthy subjects scanned twice over the same period. We studied the time by group interaction for myo-inositol after adjusting for gender and age. We report significant reduction in myo-inositol concentration in the ACC in schizophrenia at an early, untreated state of acute illness that becomes insignificant over time, after instituting early intervention. This trajectory indicates that dynamic astroglial changes are likely to operate in the early stages of schizophrenia. MRS myo-inositol may be a critical marker of amelioration of active psychosis in early stages of schizophrenia.
Collapse
Affiliation(s)
- Peter Jeon
- Department of Medical Biophysics, Western University, London, Canada
- Imaging Division, Lawson Health Research Institute, London, Canada
| | - Michael Mackinley
- Imaging Division, Lawson Health Research Institute, London, Canada
- Robarts Research Institute, Western University, London, Canada
- Department of Neuroscience, Western University, London, Canada
| | - Jean Théberge
- Department of Medical Biophysics, Western University, London, Canada
- Imaging Division, Lawson Health Research Institute, London, Canada
- Diagnostic Imaging, St. Joseph's Health Care, London, Canada
- Department of Medical Imaging, Western University, London, Canada
- Department of Psychiatry, Western University, London, Canada
| | - Lena Palaniyappan
- Department of Medical Biophysics, Western University, London, Canada.
- Imaging Division, Lawson Health Research Institute, London, Canada.
- Robarts Research Institute, Western University, London, Canada.
- Department of Psychiatry, Western University, London, Canada.
- Robarts Research Institute, UWO, 1151 Richmond Street N., Room 3208, London, ON, N6A 5B7, Canada.
| |
Collapse
|
22
|
Satheesh NJ, Salloum-Asfar S, Abdulla SA. The Potential Role of COVID-19 in the Pathogenesis of Multiple Sclerosis-A Preliminary Report. Viruses 2021; 13:2091. [PMID: 34696521 PMCID: PMC8540806 DOI: 10.3390/v13102091] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/17/2022] Open
Abstract
Coronavirus 2019 (COVID-19) is an infectious respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that mainly affects the lungs. COVID-19 symptoms include the presence of fevers, dry coughs, fatigue, sore throat, headaches, diarrhea, and a loss of taste or smell. However, it is understood that SARS-CoV-2 is neurotoxic and neuro-invasive and could enter the central nervous system (CNS) via the hematogenous route or via the peripheral nerve route and causes encephalitis, encephalopathy, and acute disseminated encephalomyelitis (ADEM) in COVID-19 patients. This review discusses the possibility of SARS-CoV-2-mediated Multiple Sclerosis (MS) development in the future, comparable to the surge in Parkinson's disease cases following the Spanish Flu in 1918. Moreover, the SARS-CoV-2 infection is associated with a cytokine storm. This review highlights the impact of these modulated cytokines on glial cell interactions within the CNS and their role in potentially prompting MS development as a secondary disease by SARS-CoV-2. SARS-CoV-2 is neurotropic and could interfere with various functions of neurons leading to MS development. The influence of neuroinflammation, microglia phagocytotic capabilities, as well as hypoxia-mediated mitochondrial dysfunction and neurodegeneration, are mechanisms that may ultimately trigger MS development.
Collapse
Affiliation(s)
| | - Salam Salloum-Asfar
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha 34110, Qatar;
| | - Sara A. Abdulla
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha 34110, Qatar;
| |
Collapse
|
23
|
Hernandez-Garcia E, Chrysikou E, Kalea AZ. The Interplay between Housing Environmental Attributes and Design Exposures and Psychoneuroimmunology Profile-An Exploratory Review and Analysis Paper in the Cancer Survivors' Mental Health Morbidity Context. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:10891. [PMID: 34682637 PMCID: PMC8536084 DOI: 10.3390/ijerph182010891] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/08/2021] [Accepted: 10/14/2021] [Indexed: 12/11/2022]
Abstract
Adult cancer survivors have an increased prevalence of mental health comorbidities and other adverse late-effects interdependent with mental illness outcomes compared with the general population. Coronavirus Disease 2019 (COVID-19) heralds an era of renewed call for actions to identify sustainable modalities to facilitate the constructs of cancer survivorship care and health care delivery through physiological supportive domestic spaces. Building on the concept of therapeutic architecture, psychoneuroimmunology (PNI) indicators-with the central role in low-grade systemic inflammation-are associated with major psychiatric disorders and late effects of post-cancer treatment. Immune disturbances might mediate the effects of environmental determinants on behaviour and mental disorders. Whilst attention is paid to the non-objective measurements for examining the home environmental domains and mental health outcomes, little is gathered about the multidimensional effects on physiological responses. This exploratory review presents a first analysis of how addressing the PNI outcomes serves as a catalyst for therapeutic housing research. We argue the crucial component of housing in supporting the sustainable primary care and public health-based cancer survivorship care model, particularly in the psychopathology context. Ultimately, we illustrate a series of interventions aiming at how housing environmental attributes can trigger PNI profile changes and discuss the potential implications in the non-pharmacological treatment of cancer survivors and patients with mental morbidities.
Collapse
Affiliation(s)
- Eva Hernandez-Garcia
- The Bartlett Real Estate Institute, The Bartlett School of Sustainable Construction, University College London, London WC1E 6BT, UK;
| | - Evangelia Chrysikou
- The Bartlett Real Estate Institute, The Bartlett School of Sustainable Construction, University College London, London WC1E 6BT, UK;
- Clinic of Social and Family Medicine, Department of Social Medicine, University of Crete, 700 13 Heraklion, Greece
| | - Anastasia Z. Kalea
- Division of Medicine, University College London, London WC1E 6JF, UK;
- Institute of Cardiovascular Science, University College London, London WC1E 6HX, UK
| |
Collapse
|
24
|
Rocha-Gomes A, Teixeira AE, de Oliveira DG, Santiago CMO, da Silva AA, Riul TR, Lacerda ACR, Mendonça VA, Rocha-Vieira E, Leite HR. LPS tolerance prevents anxiety-like behavior and amygdala inflammation of high-fat-fed dams' adolescent offspring. Behav Brain Res 2021; 411:113371. [PMID: 34019914 DOI: 10.1016/j.bbr.2021.113371] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 05/15/2021] [Accepted: 05/15/2021] [Indexed: 02/04/2023]
Abstract
Maternal high-fat diets (HFD) can generate inflammation in the offspring's amygdala, which can lead to anxiety-like behaviors. Conversely, lipopolysaccharide (LPS) tolerance can reduce neuroinflammation in the offspring caused by maternal high-fat diets. This study evaluated the combination of LPS tolerance and high-fat maternal diet on amygdala's inflammatory parameters and the anxiety-like behavior in adolescent offspring. Female pregnant Wistar rats received randomly a standard diet or a high-fat diet during gestation and lactation. On gestation days 8, 10, and 12, half of the females in each group were intraperitonially injected with LPS (0.1 mg.kg-1). After weaning, the male offspring (n = 96) were placed in individual boxes in standard conditions, and when 6 weeks-old, the animals underwent: Open-Field, Light/Dark Box, Elevated Plus-Maze, and Rotarod tests. When 50 days-old the offspring were euthanized and the amygdala removed for cytokine and redox status analysis. The offspring in the HFD group showed lower amygdala IL-10 levels, high IL-6/IL-10 ratio, and anxiety-like behaviors. These effects were attenuated in the HFD offspring submitted to LPS tolerance, which showed an anti-inflammatory compensatory response in the amygdala. Also, this group showed a higher activity of the enzyme catalase in the amygdala. In addition, receiving the combination of LPS tolerance and maternal HFD did not lead to anxiety-like behavior in the offspring. The results suggest that LPS tolerance attenuated amygdala inflammation through an anti-inflammatory compensatory response besides preventing anxiety-like behavior caused by the high-fat maternal diet.
Collapse
Affiliation(s)
- Arthur Rocha-Gomes
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil; Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil
| | - Amanda Escobar Teixeira
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil
| | - Dalila Gomes de Oliveira
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil
| | - Camilla Mainy Oliveira Santiago
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil; Programa de Pós-Graduação em Ciências da Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil
| | - Alexandre Alves da Silva
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil
| | - Tania Regina Riul
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil; Programa de Pós-Graduação em Ciências da Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil
| | - Ana Cristina Rodrigues Lacerda
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil
| | - Vanessa Amaral Mendonça
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil
| | - Etel Rocha-Vieira
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil; Faculdade de Medicina do Campus JK, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil
| | - Hércules Ribeiro Leite
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil; Programa de Pós Graduação em Ciências da Reabilitação, Universidade Federal de Minas Gerais, Diamantina, MG, Brazil.
| |
Collapse
|
25
|
Gammon D, Cheng C, Volkovinskaia A, Baker GB, Dursun SM. Clozapine: Why Is It So Uniquely Effective in the Treatment of a Range of Neuropsychiatric Disorders? Biomolecules 2021; 11:1030. [PMID: 34356654 PMCID: PMC8301879 DOI: 10.3390/biom11071030] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/30/2021] [Accepted: 07/02/2021] [Indexed: 12/16/2022] Open
Abstract
Clozapine is superior to other antipsychotics as a therapy for treatment-resistant schizophrenia and schizoaffective disorder with increased risk of suicidal behavior. This drug has also been used in the off-label treatment of bipolar disorder, major depressive disorder (MDD), and Parkinson's disease (PD). Although usually reserved for severe and treatment-refractory cases, it is interesting that electroconvulsive therapy (ECT) has also been used in the treatment of these psychiatric disorders, suggesting some common or related mechanisms. A literature review on the applications of clozapine and electroconvulsive therapy (ECT) to the disorders mentioned above was undertaken, and this narrative review was prepared. Although both treatments have multiple actions, evidence to date suggests that the ability to elicit epileptiform activity and alter EEG activity, to increase neuroplasticity and elevate brain levels of neurotrophic factors, to affect imbalances in the relationship between glutamate and γ-aminobutyric acid (GABA), and to reduce inflammation through effects on neuron-glia interactions are common underlying mechanisms of these two treatments. This evidence may explain why clozapine is effective in a range of neuropsychiatric disorders. Future increased investigations into epigenetic and connectomic changes produced by clozapine and ECT should provide valuable information about these two treatments and the disorders they are used to treat.
Collapse
Affiliation(s)
- Dara Gammon
- Saba University School of Medicine, Saba, The Netherlands; (D.G.); (A.V.)
| | - Catherine Cheng
- Neurochemical Research Unit and Bebensee Schizophrenia Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB T6G 2B7, Canada; (C.C.); (G.B.B.)
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada
| | - Anna Volkovinskaia
- Saba University School of Medicine, Saba, The Netherlands; (D.G.); (A.V.)
| | - Glen B. Baker
- Neurochemical Research Unit and Bebensee Schizophrenia Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB T6G 2B7, Canada; (C.C.); (G.B.B.)
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Serdar M. Dursun
- Neurochemical Research Unit and Bebensee Schizophrenia Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB T6G 2B7, Canada; (C.C.); (G.B.B.)
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada
| |
Collapse
|
26
|
Altaf-Ul-Amin M, Hirose K, Nani JV, Porta LC, Tasic L, Hossain SF, Huang M, Ono N, Hayashi MAF, Kanaya S. A system biology approach based on metabolic biomarkers and protein-protein interactions for identifying pathways underlying schizophrenia and bipolar disorder. Sci Rep 2021; 11:14450. [PMID: 34262063 PMCID: PMC8280132 DOI: 10.1038/s41598-021-93653-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 06/28/2021] [Indexed: 11/10/2022] Open
Abstract
Mental disorders (MDs), including schizophrenia (SCZ) and bipolar disorder (BD), have attracted special attention from scientists due to their high prevalence and significantly debilitating clinical features. The diagnosis of MDs is still essentially based on clinical interviews, and intensive efforts to introduce biochemical based diagnostic methods have faced several difficulties for implementation in clinics, due to the complexity and still limited knowledge in MDs. In this context, aiming for improving the knowledge in etiology and pathophysiology, many authors have reported several alterations in metabolites in MDs and other brain diseases. After potentially fishing all metabolite biomarkers reported up to now for SCZ and BD, we investigated here the proteins related to these metabolites in order to construct a protein-protein interaction (PPI) network associated with these diseases. We determined the statistically significant clusters in this PPI network and, based on these clusters, we identified 28 significant pathways for SCZ and BDs that essentially compose three groups representing three major systems, namely stress response, energy and neuron systems. By characterizing new pathways with potential to innovate the diagnosis and treatment of psychiatric diseases, the present data may also contribute to the proposal of new intervention for the treatment of still unmet aspects in MDs.
Collapse
Affiliation(s)
- Md Altaf-Ul-Amin
- Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan.
| | - Kazuhisa Hirose
- Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - João V Nani
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
- National Institute for Translational Medicine (INCT-TM, CNPq/FAPESP/CAPES), Ribeirão Preto, Brazil
| | - Lucas C Porta
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Ljubica Tasic
- Chemical Biology Laboratory, Department of Organic Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (Unicamp), Campinas, SP, Brazil
| | | | - Ming Huang
- Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Naoaki Ono
- Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Mirian A F Hayashi
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil.
- National Institute for Translational Medicine (INCT-TM, CNPq/FAPESP/CAPES), Ribeirão Preto, Brazil.
| | - Shigehiko Kanaya
- Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| |
Collapse
|
27
|
Recordati C, De Maglie M, Cella C, Argentiere S, Paltrinieri S, Bianchessi S, Losa M, Fiordaliso F, Corbelli A, Milite G, Aureli F, D’Amato M, Raggi A, Cubadda F, Soldati S, Lenardi C, Scanziani E. Repeated oral administration of low doses of silver in mice: tissue distribution and effects on central nervous system. Part Fibre Toxicol 2021; 18:23. [PMID: 34134756 PMCID: PMC8207582 DOI: 10.1186/s12989-021-00418-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 06/02/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Widespread use of silver in its different forms raises concerns about potential adverse effects after ingestion, the main exposure route for humans. The aim of this study was to investigate in CD-1 (ICR) male mice the tissue distribution and in vivo effects of 4-week oral exposure to 0.25 and 1 mg Ag/kg bw 10 nm citrate coated silver nanoparticles (AgNPs) and 1 mg Ag/kg bw silver acetate (AgAc) at the end of treatment (EoT) and after 4 weeks of recovery. RESULTS There were no treatment-related clinical signs and mortality, and no significant effects on body and organ weights at the EoT and after recovery. Treatment-related changes in hematology and clinical chemistry were found after recovery, the most relevant being a dose-dependent lymphopenia and increased triglycerides in AgNP-treated mice, and increased levels of urea in all treated groups, associated with decreased albumin only in AgAc-treated mice. At the EoT the highest silver concentration determined by Triple Quadrupole ICP-MS analysis was found in the brain, followed by testis, liver, and spleen; much lower concentrations were present in the small intestine and kidney. Tissue silver concentrations were slightly higher after exposure to AgAc than AgNPs and dose dependent for AgNPs. After recovery silver was still present in the brain and testis, highlighting slow elimination. No histopathological changes and absence of silver staining by autometallography were observed in the organs of treated mice. At the EoT GFAP (astrocytes) immunoreactivity was significantly increased in the hippocampus of AgNP-treated mice in a dose-dependent manner and Iba1 (microglial cells) immunoreactivity was significantly increased in the cortex of 1 mg/kg bw AgNP-treated mice. After recovery, a significant reduction of Iba1 was observed in the cortex of all treated groups. TEM analysis of the hippocampus revealed splitting of basement membrane of the capillaries and swelling of astrocytic perivascular end-feet in 1 mg/kg bw AgNP- and AgAc-treated mice at the EoT. CONCLUSIONS Our study revealed accumulation and slow clearance of silver in the brain after oral administration of 10 nm AgNPs and AgAc at low doses in mice, associated with effects on glial cells and ultrastructural alterations of the Blood-Brain Barrier.
Collapse
Affiliation(s)
- Camilla Recordati
- Dipartimento di Medicina Veterinaria (DIMEVET), Università degli Studi di Milano, 26900 Lodi, Italy
- Fondazione Unimi, 20139 Milan, Italy
| | - Marcella De Maglie
- Dipartimento di Medicina Veterinaria (DIMEVET), Università degli Studi di Milano, 26900 Lodi, Italy
- Fondazione Unimi, 20139 Milan, Italy
| | - Claudia Cella
- Fondazione Unimi, 20139 Milan, Italy
- Dipartimento di Fisica, Università degli Studi di Milano, 20133 Milan, Italy
| | - Simona Argentiere
- Fondazione Unimi, 20139 Milan, Italy
- Dipartimento di Fisica, Università degli Studi di Milano, 20133 Milan, Italy
| | - Saverio Paltrinieri
- Dipartimento di Medicina Veterinaria (DIMEVET), Università degli Studi di Milano, 26900 Lodi, Italy
| | | | | | - Fabio Fiordaliso
- Unit of Bio-imaging, Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Alessandro Corbelli
- Unit of Bio-imaging, Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | | | - Federica Aureli
- Istituto Superiore di Sanità - National Institute of Health, 00161 Rome, Italy
| | - Marilena D’Amato
- Istituto Superiore di Sanità - National Institute of Health, 00161 Rome, Italy
| | - Andrea Raggi
- Istituto Superiore di Sanità - National Institute of Health, 00161 Rome, Italy
| | - Francesco Cubadda
- Istituto Superiore di Sanità - National Institute of Health, 00161 Rome, Italy
| | | | - Cristina Lenardi
- Fondazione Unimi, 20139 Milan, Italy
- Dipartimento di Fisica, Università degli Studi di Milano, 20133 Milan, Italy
- Centro Interdisciplinare Materiali e Interfacce Nanostrutturati (CIMAINA), Università degli Studi di Milano, 20133 Milan, Italy
| | - Eugenio Scanziani
- Dipartimento di Medicina Veterinaria (DIMEVET), Università degli Studi di Milano, 26900 Lodi, Italy
- Fondazione Unimi, 20139 Milan, Italy
| |
Collapse
|
28
|
Rusinek K, Sołek P, Tabęcka-Łonczyńska A, Koziorowski M, Mytych J. Focus on the Role of Klotho Protein in Neuro-Immune Interactions in HT-22 Cells Upon LPS Stimulation. Cells 2020; 9:E1231. [PMID: 32429346 PMCID: PMC7290853 DOI: 10.3390/cells9051231] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/07/2020] [Accepted: 05/15/2020] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation is defined as the activation of the brain's innate immune system in response to an inflammatory challenge and is considered to be a prominent feature of neurodegenerative diseases. The contribution of overactivated neuroglial cells to neuroinflammation and neurodegenerative disorders is well documented, however, the role of hippocampal neurons in the neuroinflammatory process remains fragmentary. In this study, we show for the first time, that klotho acts as a signal transducer between pro-survival and pro-apoptotic crosstalk mediated by ER stress in HT-22 hippocampal neuronal cells during LPS challenge. In control HT-22 cells, LPS treatment results in activation of the IRE1α-p38 MAPK pathway leading to increased secretion of anti-inflammatory IL-10, and thus, providing adaptation mechanism. On the other hand, in klotho-deficient HT-22 cells, LPS induces oxi-nitrosative stress and genomic instability associated with telomere dysfunctions leading to p53/p21-mediated cell cycle arrest and, in consequence, to ER stress, inflammation as well as of apoptotic cell death. Therefore, these results indicate that klotho serves as a part of the cellular defense mechanism engaged in the protection of neuronal cells against LPS-mediated neuroinflammation, emerging issues linked with neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | | | | | - Jennifer Mytych
- Department of Animal Physiology and Reproduction, Institute of Biology and Biotechnology, Collegium Scientarium Naturalium, University of Rzeszow, Werynia 2, 36-100 Kolbuszowa, Poland; (K.R.); (P.S.); (A.T.-Ł.); (M.K.)
| |
Collapse
|