1
|
Razavi SZ, Amini-Khoei H, Rahimi-Madiseh M, Bijad E, Lorigooini Z. Modulation of neuroinflammation and oxidative stress by Echinacea purpurea extract: Therapeutic potential in maternal separation-induced autism spectrum disorder. J Psychiatr Res 2025; 184:118-127. [PMID: 40049118 DOI: 10.1016/j.jpsychires.2025.02.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 06/21/2024] [Accepted: 02/20/2025] [Indexed: 04/09/2025]
Abstract
BACKGROUND Autism Spectrum Disorder (ASD) is defined by ongoing problems in social interaction and communication and repetitive, constrained behavior patterns. The link between oxidative stress (OS) and inflammation with ASD has been shown in previous studies. E. purpurea is well-known for its potential antioxidant and anti-inflammatory pharmacological properties. In this study, we aimed to evaluate the effects of E. purpurea hydroalcoholic extract on autistic-like behaviors following a mouse model of maternal separation (MS) stress, focusing on possible anti-neuroinflammation and antioxidative stress. METHODS 70% hydro-ethanolic extract was macerated from the aerial parts of E. purpurea. Standardization was done by determining the amount of chicoric acid in the extract using the UHPLC method. Then, behavioral analysis was done on 75 male mice that underwent MS. Mice were treated with normal saline or 75, 150, and 300 mg/kg of the extract. Sociability behaviors and stereotyping behaviors have been evaluated. Also, their total antioxidant capacity (TAC), nitrite levels, and malondialdehyde (MDA) were measured in the hippocampus. In addition, the expression of inflammatory factors, including interleukin-1 (IL-1), NLRP3, and TLR4, has been determined by quantitative real-time PCR (qRT-PCR). Data were analyzed after collection using PRISM statistical software. RESULTS Our findings indicated that MS caused autistic-like behaviors in mice (increased sociability index and social preference index) and increased repetitive behaviors (increased number of buried marbles). These autistic-like behaviors are associated with increased MDA, nitrite, over-expression of inflammatory genes, decreased MDA, nitrite, over-expression of inflammatory genes, and decreased TAC in the hippocampus. E. purpurea extract significantly reversed these adverse effects of MS. CONCLUSION The results of this study showed that E. purpurea extract might reduce autistic-like behaviors in MS by attenuating neuroinflammation and oxidative stress states.
Collapse
Affiliation(s)
- Seyedeh Zahra Razavi
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hossein Amini-Khoei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad Rahimi-Madiseh
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Elham Bijad
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zahra Lorigooini
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
2
|
Sleiman A, Miller KB, Flores D, Kuan J, Altwasser K, Smith BJ, Kozbenko T, Hocking R, Wood SJ, Huff J, Adam-Guillermin C, Hamada N, Yauk C, Wilkins R, Chauhan V. AOP report: Development of an adverse outcome pathway for deposition of energy leading to learning and memory impairment. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2024; 65 Suppl 3:57-84. [PMID: 39228295 DOI: 10.1002/em.22622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/05/2024]
Abstract
Understanding radiation-induced non-cancer effects on the central nervous system (CNS) is essential for the risk assessment of medical (e.g., radiotherapy) and occupational (e.g., nuclear workers and astronauts) exposures. Herein, the adverse outcome pathway (AOP) approach was used to consolidate relevant studies in the area of cognitive decline for identification of research gaps, countermeasure development, and for eventual use in risk assessments. AOPs are an analytical construct describing critical events to an adverse outcome (AO) in a simplified form beginning with a molecular initiating event (MIE). An AOP was constructed utilizing mechanistic information to build empirical support for the key event relationships (KERs) between the MIE of deposition of energy to the AO of learning and memory impairment through multiple key events (KEs). The evidence for the AOP was acquired through a documented scoping review of the literature. In this AOP, the MIE is connected to the AO via six KEs: increased oxidative stress, increased deoxyribonucleic acid (DNA) strand breaks, altered stress response signaling, tissue resident cell activation, increased pro-inflammatory mediators, and abnormal neural remodeling that encompasses atypical structural and functional alterations of neural cells and surrounding environment. Deposition of energy directly leads to oxidative stress, increased DNA strand breaks, an increase of pro-inflammatory mediators and tissue resident cell activation. These KEs, which are themselves interconnected, can lead to abnormal neural remodeling impacting learning and memory processes. Identified knowledge gaps include improving quantitative understanding of the AOP across several KERs and additional testing of proposed modulating factors through experimental work. Broadly, it is envisioned that the outcome of these efforts could be extended to other cognitive disorders and complement ongoing work by international radiation governing bodies in their review of the system of radiological protection.
Collapse
Affiliation(s)
- Ahmad Sleiman
- Institut de Radioprotection et de Sûreté Nucléaire, St. Paul Lez Durance, Provence, France
| | - Kathleen B Miller
- Department of Health and Exercise Science, Morrison College Family of Health, University of St. Thomas, Saint Paul, Minnesota, USA
| | - Danicia Flores
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Jaqueline Kuan
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Kaitlyn Altwasser
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Benjamin J Smith
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Tatiana Kozbenko
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Robyn Hocking
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | | | - Janice Huff
- NASA Langley Research Center, Hampton, Virginia, USA
| | | | - Nobuyuki Hamada
- Biology and Environmental Chemistry Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Chiba, Japan
| | - Carole Yauk
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Ruth Wilkins
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Vinita Chauhan
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| |
Collapse
|
3
|
Albekairi TH, Alanazi MM, Ansari MA, Nadeem A, Attia SM, Bakheet SA, Al-Mazroua HA, Aldossari AA, Almanaa TN, Alwetaid MY, Alqinyah M, Alnefaie HO, Ahmad SF. Cadmium exposure exacerbates immunological abnormalities in a BTBR T + Itpr3 tf/J autistic mouse model by upregulating inflammatory mediators in CD45R-expressing cells. J Neuroimmunol 2024; 386:578253. [PMID: 38064869 DOI: 10.1016/j.jneuroim.2023.578253] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 11/26/2023] [Accepted: 11/29/2023] [Indexed: 01/13/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental illness characterized by behavior, learning, communication, and social interaction abnormalities in various situations. Individuals with impairments usually exhibit restricted and repetitive actions. The actual cause of ASD is yet unknown. It is believed, however, that a mix of genetic and environmental factors may play a role in its development. Certain metals have been linked to the development of neurological diseases, and the prevalence of ASD has shown a positive association with industrialization. Cadmium chloride (Cd) is a neurotoxic chemical linked to cognitive impairment, tremors, and neurodegenerative diseases. The BTBR T+ Itpr3tf/J (BTBR) inbred mice are generally used as a model for ASD and display a range of autistic phenotypes. We looked at how Cd exposure affected the signaling of inflammatory mediators in CD45R-expressing cells in the BTBR mouse model of ASD. In this study, we looked at how Cd affected the expression of numerous markers in the spleen, including IFN-γ, IL-6, NF-κB p65, GM-CSF, iNOS, MCP-1, and Notch1. Furthermore, we investigated the effect of Cd exposure on the expression levels of numerous mRNA molecules in brain tissue, including IFN-γ, IL-6, NF-κB p65, GM-CSF, iNOS, MCP-1, and Notch1. The RT-PCR technique was used for this analysis. Cd exposure increased the number of CD45R+IFN-γ+, CD45R+IL-6+, CD45R+NF-κB p65+, CD45R+GM-CSF+, CD45R+GM-CSF+, CD45R+iNOS+, and CD45R+Notch1+ cells in the spleen of BTBR mice. Cd treatment also enhanced mRNA expression in brain tissue for IFN-γ, IL-6, NF-κB, GM-CSF, iNOS, MCP-1, and Notch1. In general, Cd increases the signaling of inflammatory mediators in BTBR mice. This study is the first to show that Cd exposure causes immune function dysregulation in the BTBR ASD mouse model. As a result, our study supports the role of Cd exposure in the development of ASD.
Collapse
Affiliation(s)
- Thamer H Albekairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed M Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Haneen A Al-Mazroua
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah A Aldossari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Taghreed N Almanaa
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammad Y Alwetaid
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed Alqinyah
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Hajar O Alnefaie
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
4
|
Molecular Imaging of Ultrasound-Mediated Blood-Brain Barrier Disruption in a Mouse Orthotopic Glioblastoma Model. Pharmaceutics 2022; 14:pharmaceutics14102227. [PMID: 36297663 PMCID: PMC9610067 DOI: 10.3390/pharmaceutics14102227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022] Open
Abstract
Glioblastoma (GBM) is an aggressive and malignant primary brain tumor. The blood-brain barrier (BBB) limits the therapeutic options available to tackle this incurable tumor. Transient disruption of the BBB by focused ultrasound (FUS) is a promising and safe approach to increase the brain and tumor concentration of drugs administered systemically. Non-invasive, sensitive, and reliable imaging approaches are required to better understand the impact of FUS on the BBB and brain microenvironment. In this study, nuclear imaging (SPECT/CT and PET/CT) was used to quantify neuroinflammation 48 h post-FUS and estimate the influence of FUS on BBB opening and tumor growth in vivo. BBB disruptions were performed on healthy and GBM-bearing mice (U-87 MG xenograft orthotopic model). The BBB recovery kinetics were followed and quantified by [99mTc]Tc-DTPA SPECT/CT imaging at 0.5 h, 3 h and 24 h post-FUS. The absence of neuroinflammation was confirmed by [18F]FDG PET/CT imaging 48 h post-FUS. The presence of the tumor and its growth were evaluated by [68Ga]Ga-RGD2 PET/CT imaging and post-mortem histological analysis, showing that tumor growth was not influenced by FUS. In conclusion, molecular imaging can be used to evaluate the time frame for systemic treatment combined with transient BBB opening and to test its efficacy over time.
Collapse
|
5
|
Díaz HS, Ríos-Gallardo A, Ortolani D, Díaz-Jara E, Flores MJ, Vera I, Monasterio A, Ortiz FC, Brossard N, Osorio F, Río RD. Lipid-Encapsuled Grape Tannins Prevent Oxidative-Stress-Induced Neuronal Cell Death, Intracellular ROS Accumulation and Inflammation. Antioxidants (Basel) 2022; 11:antiox11101928. [PMID: 36290649 PMCID: PMC9598423 DOI: 10.3390/antiox11101928] [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: 08/01/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/16/2022] Open
Abstract
The central nervous system (CNS) is particularly vulnerable to oxidative stress and inflammation, which affect neuronal function and survival. Nowadays, there is great interest in the development of antioxidant and anti-inflammatory compounds extracted from natural products, as potential strategies to reduce the oxidative/inflammatory environment within the CNS and then preserve neuronal integrity and brain function. However, an important limitation of natural antioxidant formulations (mainly polyphenols) is their reduced in vivo bioavailability. The biological compatible delivery system containing polyphenols may serve as a novel compound for these antioxidant formulations. Accordingly, in the present study, we used liposomes as carriers for grape tannins, and we tested their ability to prevent neuronal oxidative stress and inflammation. Cultured catecholaminergic neurons (CAD) were used to establish the potential of lipid-encapsulated grape tannins (TLS) to prevent neuronal oxidative stress and inflammation following an oxidative insult. TLS rescued cell survival after H2O2 treatment (59.4 ± 8.8% vs. 90.4 ± 5.6% H2O2 vs. TLS+ H2O2; p < 0.05) and reduced intracellular ROS levels by ~38% (p < 0.05), despite displaying negligible antioxidant activity in solution. Additionally, TLS treatment dramatically reduced proinflammatory cytokines’ mRNA expression after H2O2 treatment (TNF-α: 400.3 ± 1.7 vs. 7.9 ± 1.9-fold; IL-1β: 423.4 ± 1.3 vs. 12.7 ± 2.6-fold; p < 0.05; H2O2 vs. TLS+ H2O2, respectively), without affecting pro/antioxidant biomarker expression, suggesting that liposomes efficiently delivered tannins inside neurons and promoted cell survival. In conclusion, we propose that lipid-encapsulated grape tannins could be an efficient tool to promote antioxidant/inflammatory cell defense.
Collapse
Affiliation(s)
- Hugo S. Díaz
- Laboratory of Cardiorespiratory Control, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
| | - Angélica Ríos-Gallardo
- Laboratory of Cardiorespiratory Control, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
| | - Domiziana Ortolani
- Laboratory of Cardiorespiratory Control, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
| | - Esteban Díaz-Jara
- Laboratory of Cardiorespiratory Control, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
| | - María José Flores
- Laboratory of Cardiorespiratory Control, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
| | - Ignacio Vera
- Laboratory of Cardiorespiratory Control, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
| | - Angela Monasterio
- Departamento de Ciencia y Tecnología de Alimentos, Facultad Tecnológica, Universidad de Santiago de Chile, Santiago 8320000, Chile
| | - Fernando C. Ortiz
- Mechanisms of Myelin Formation and Repair Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 8320000, Chile
| | - Natalia Brossard
- Department of Fruit Production and Enology, School of Agricultural and Forest Sciences, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
| | - Fernando Osorio
- Departamento de Ciencia y Tecnología de Alimentos, Facultad Tecnológica, Universidad de Santiago de Chile, Santiago 8320000, Chile
| | - Rodrigo Del Río
- Laboratory of Cardiorespiratory Control, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
- Centro de Envejecimiento y Regeneración CARE-UC, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas 6200000, Chile
- Correspondence:
| |
Collapse
|
6
|
Mudra Rakshasa-Loots A, Whalley HC, Vera JH, Cox SR. Neuroinflammation in HIV-associated depression: evidence and future perspectives. Mol Psychiatry 2022; 27:3619-3632. [PMID: 35618889 PMCID: PMC9708589 DOI: 10.1038/s41380-022-01619-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/04/2022] [Accepted: 05/10/2022] [Indexed: 02/08/2023]
Abstract
People living with HIV face a high risk of mental illness, especially depression. We do not yet know the precise neurobiological mechanisms underlying HIV-associated depression. Depression severity in the general population has been linked to acute and chronic markers of systemic inflammation. Given the associations between depression and peripheral inflammation, and since HIV infection in the brain elicits a neuroinflammatory response, it is possible that neuroinflammation contributes to the high prevalence of depression amongst people living with HIV. The purpose of this review was to synthesise existing evidence for associations between inflammation, depression, and HIV. While there is strong evidence for independent associations between these three conditions, few preclinical or clinical studies have attempted to characterise their interrelationship, representing a major gap in the literature. This review identifies key areas of debate in the field and offers perspectives for future investigations of the pathophysiology of HIV-associated depression. Reproducing findings across diverse populations will be crucial in obtaining robust and generalisable results to elucidate the precise role of neuroinflammation in this pathophysiology.
Collapse
Affiliation(s)
- Arish Mudra Rakshasa-Loots
- Edinburgh Neuroscience, School of Biomedical Sciences, The University of Edinburgh, Edinburgh, UK.
- Lothian Birth Cohorts Group, Department of Psychology, The University of Edinburgh, Edinburgh, UK.
| | - Heather C Whalley
- Division of Psychiatry, Centre for Clinical Brain Sciences, Royal Edinburgh Hospital, The University of Edinburgh, Edinburgh, UK
| | - Jaime H Vera
- Department of Global Health and Infection, Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | - Simon R Cox
- Lothian Birth Cohorts Group, Department of Psychology, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
7
|
Manjeese W, Mvubu NE, Steyn AJC, Mpofana T. Mycobacterium tuberculosis causes a leaky blood-brain barrier and neuroinflammation in the prefrontal cortex and cerebellum regions of infected mice offspring. Int J Dev Neurosci 2021; 81:428-437. [PMID: 33932039 DOI: 10.1002/jdn.10116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/17/2021] [Accepted: 04/25/2021] [Indexed: 12/18/2022] Open
Abstract
The maternal system's exposure to pathogens influences foetal brain development through the influx of maternal cytokines and activation of the foetal immune status to a persistent inflammatory state characterised by glia cell activation. Neuroinflammation influences the blood-brain barrier's (BBB) permeability allowing peripheral immune cell trafficking into the brain. Mycobacterium tuberculosis (Mtb) is a pathogen that causes Tuberculosis (TB), a global pandemic responsible for health and economic burdens. Although it is known that maternal infections increase the risk of Autism spectrum disorder (ASD), it is not known whether gestational Mtb infections also contribute to impaired foetal neurodevelopment. Here we infect pregnant Balb/c mice with Mtb H37Rv and Valproic acid (VPA) individually and in combination. Neuroinflammation was measured by assessing microglia and astrocyte population in the prefrontal cortex (PFC) and cerebellum (CER) of pups. Mtb infection increased the microglia population and caused morphological changes to a reactive phenotype in the PFC. Also, the astrocyte population was significantly increased in the PFC of Mtb pups. The BBB permeability was determined by measuring the Evans Blue (EB) dye concentration in the PFC and CER 1 hr post receiving intravenous EB-dye injection. We found that prenatal Mtb exposure significantly increased the BBB's permeability in the PFC and CER of pups versus saline. Overall, our data demonstrate that prenatal exposure to Mtb predisposes offspring to a higher risk of BBB damage while inducing persistent neuroinflammation, which could lead to impaired neuronal development and function. These findings implicate a potential role of gestational Mtb infections in the aetiology of ASD.
Collapse
Affiliation(s)
- Wadzanai Manjeese
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of Kwazulu Natal, Durban, South Africa
| | - Nontobeko E Mvubu
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of Kwazulu Natal, Durban, South Africa
| | - Adrie J C Steyn
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of Kwazulu Natal, Durban, South Africa.,Africa Health Research Institute, K-Rith Tower Building, Nelson Mandela School of Medicine, Durban, South Africa.,Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Thabisile Mpofana
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of Kwazulu Natal, Durban, South Africa
| |
Collapse
|
8
|
Indika NLR, Deutz NEP, Engelen MPKJ, Peiris H, Wijetunge S, Perera R. Sulfur amino acid metabolism and related metabotypes of autism spectrum disorder: A review of biochemical evidence for a hypothesis. Biochimie 2021; 184:143-157. [PMID: 33675854 DOI: 10.1016/j.biochi.2021.02.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 02/07/2023]
Abstract
There are multiple lines of evidence for an impaired sulfur amino acid (SAA) metabolism in autism spectrum disorder (ASD). For instance, the concentrations of methionine, cysteine and S-adenosylmethionine (SAM) in body fluids of individuals with ASD is significantly lower while the concentration of S-adenosylhomocysteine (SAH) is significantly higher as compared to healthy individuals. Reduced methionine and SAM may reflect impaired remethylation pathway whereas increased SAH may reflect reduced S-adenosylhomocysteine hydrolase activity in the catabolic direction. Reduced SAM/SAH ratio reflects an impaired methylation capacity. We hypothesize multiple mechanisms to explain how the interplay of oxidative stress, neuroinflammation, mercury exposure, maternal use of valproate, altered gut microbiome and certain genetic variants may lead to these SAA metabotypes. Furthermore, we also propose a number of mechanisms to explain the metabolic consequences of abnormal SAA metabotypes. For instance in the brain, reduced SAM/SAH ratio will result in melatonin deficiency and hypomethylation of a number of biomolecules such as DNA, RNA and histones. In addition to previously proposed mechanisms, we propose that impaired activity of "radical SAM" enzymes will result in reduced endogenous lipoic acid synthesis, reduced molybdenum cofactor synthesis and impaired porphyrin metabolism leading to mitochondrial dysfunction, porphyrinuria and impaired sulfation capacity. Furthermore depletion of SAM may also lead to the disturbed mTOR signaling pathway in a subgroup of ASD. The proposed "SAM-depletion hypothesis" is an inclusive model to explain the relationship between heterogeneous risk factors and metabotypes observed in a subset of children with ASD.
Collapse
Affiliation(s)
- Neluwa-Liyanage R Indika
- Department of Biochemistry, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, Sri Lanka.
| | - Nicolaas E P Deutz
- Center for Translational Research in Aging & Longevity, Department of Health & Kinesiology, Texas A&M University, College Station, TX, USA
| | - Marielle P K J Engelen
- Center for Translational Research in Aging & Longevity, Department of Health & Kinesiology, Texas A&M University, College Station, TX, USA
| | - Hemantha Peiris
- Department of Biochemistry, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Swarna Wijetunge
- Child and Adolescent Mental Health Service, Lady Ridgeway Hospital for Children, Colombo 8, Sri Lanka
| | - Rasika Perera
- Department of Biochemistry, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| |
Collapse
|
9
|
Dang J, Tiwari SK, Agrawal K, Hui H, Qin Y, Rana TM. Glial cell diversity and methamphetamine-induced neuroinflammation in human cerebral organoids. Mol Psychiatry 2021; 26:1194-1207. [PMID: 32051547 PMCID: PMC7423603 DOI: 10.1038/s41380-020-0676-x] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 12/03/2019] [Accepted: 01/30/2020] [Indexed: 11/26/2022]
Abstract
Methamphetamine (METH) is a potent stimulant that induces a euphoric state but also causes cognitive impairment, neurotoxicity and neurodevelopmental deficits. Yet, the molecular mechanisms by which METH causes neurodevelopmental defects have remained elusive. Here we utilized human cerebral organoids and single-cell RNA sequencing (scRNA-seq) to study the effects of prenatal METH exposure on fetal brain development. We analyzed 20,758 cells from eight untreated and six METH-treated cerebral organoids and found that the organoids developed from embryonic stem cells contained a diverse array of glial and neuronal cell types. We further identified transcriptionally distinct populations of astrocytes and oligodendrocytes within cerebral organoids. Treatment of organoids with METH-induced marked changes in transcription in multiple cell types, including astrocytes and neural progenitor cells. METH also elicited novel astrocyte-specific gene expression networks regulating responses to cytokines, and inflammasome. Moreover, upregulation of immediate early genes, complement factors, apoptosis, and immune response genes suggests a neuroinflammatory program induced by METH regulating neural stem cell proliferation, differentiation, and cell death. Finally, we observed marked METH-induced changes in neuroinflammatory and cytokine gene expression at the RNA and protein levels. Our data suggest that human cerebral organoids represent a model system to study drug-induced neuroinflammation at single-cell resolution.
Collapse
Affiliation(s)
- Jason Dang
- grid.266100.30000 0001 2107 4242Division of Genetics, Department of Pediatrics, Institute for Genomic Medicine, Program in Immunology, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, CA 92093 USA
| | - Shashi Kant Tiwari
- grid.266100.30000 0001 2107 4242Division of Genetics, Department of Pediatrics, Institute for Genomic Medicine, Program in Immunology, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, CA 92093 USA
| | - Kriti Agrawal
- grid.266100.30000 0001 2107 4242Division of Genetics, Department of Pediatrics, Institute for Genomic Medicine, Program in Immunology, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, CA 92093 USA ,grid.266100.30000 0001 2107 4242Department of Biology, Bioinformatics Program, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, CA 92093 USA
| | - Hui Hui
- grid.266100.30000 0001 2107 4242Division of Genetics, Department of Pediatrics, Institute for Genomic Medicine, Program in Immunology, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, CA 92093 USA ,grid.266100.30000 0001 2107 4242Department of Biology, Bioinformatics Program, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, CA 92093 USA
| | - Yue Qin
- grid.266100.30000 0001 2107 4242Division of Genetics, Department of Pediatrics, Institute for Genomic Medicine, Program in Immunology, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, CA 92093 USA ,grid.266100.30000 0001 2107 4242Department of Biology, Bioinformatics Program, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, CA 92093 USA
| | - Tariq M. Rana
- grid.266100.30000 0001 2107 4242Division of Genetics, Department of Pediatrics, Institute for Genomic Medicine, Program in Immunology, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, CA 92093 USA
| |
Collapse
|
10
|
Eissa N, Sadeq A, Sasse A, Sadek B. Role of Neuroinflammation in Autism Spectrum Disorder and the Emergence of Brain Histaminergic System. Lessons Also for BPSD? Front Pharmacol 2020; 11:886. [PMID: 32612529 PMCID: PMC7309953 DOI: 10.3389/fphar.2020.00886] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 05/29/2020] [Indexed: 12/27/2022] Open
Abstract
Many behavioral and psychological symptoms of dementia (BPSD) share similarities in executive functioning and communication deficits with those described in several neuropsychiatric disorders, including Alzheimer's disease (AD), epilepsy, schizophrenia (SCH), and autism spectrum disorder (ASD). Numerous studies over the last four decades have documented altered neuroinflammation among individuals diagnosed with ASD. The purpose of this review is to examine the hypothesis that central histamine (HA) plays a significant role in the regulation of neuroinflammatory processes of microglia functions in numerous neuropsychiatric diseases, i.e., ASD, AD, SCH, and BPSD. In addition, this review summarizes the latest preclinical and clinical results that support the relevance of histamine H1-, H2-, and H3-receptor antagonists for the potential clinical use in ASD, SCH, AD, epilepsy, and BPSD, based on the substantial symptomatic overlap between these disorders with regards to cognitive dysfunction. The review focuses on the histaminergic neurotransmission as relevant in these brain disorders, as well as the effects of a variety of H3R antagonists in animal models and in clinical studies.
Collapse
Affiliation(s)
- Nermin Eissa
- Department of Pharmacology and Therapeutics, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates.,Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Adel Sadeq
- College of Pharmacy, Al Ain University of Science and Technology, Al Ain, United Arab Emirates
| | - Astrid Sasse
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, University of Dublin, Dublin, Ireland
| | - Bassem Sadek
- Department of Pharmacology and Therapeutics, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates.,Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
11
|
Alvarez-Carbonell D, Ye F, Ramanath N, Garcia-Mesa Y, Knapp PE, Hauser KF, Karn J. Cross-talk between microglia and neurons regulates HIV latency. PLoS Pathog 2019; 15:e1008249. [PMID: 31887215 PMCID: PMC6953890 DOI: 10.1371/journal.ppat.1008249] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 01/10/2020] [Accepted: 12/01/2019] [Indexed: 12/30/2022] Open
Abstract
Despite effective antiretroviral therapy (ART), HIV-associated neurocognitive disorders (HAND) are found in nearly one-third of patients. Using a cellular co-culture system including neurons and human microglia infected with HIV (hμglia/HIV), we investigated the hypothesis that HIV-dependent neurological degeneration results from the periodic emergence of HIV from latency within microglial cells in response to neuronal damage or inflammatory signals. When a clonal hμglia/HIV population (HC69) expressing HIV, or HIV infected human primary and iPSC-derived microglial cells, were cultured for a short-term (24 h) with healthy neurons, HIV was silenced. The neuron-dependent induction of latency in HC69 cells was recapitulated using induced pluripotent stem cell (iPSC)-derived GABAergic cortical (iCort) and dopaminergic (iDopaNer), but not motor (iMotorNer), neurons. By contrast, damaged neurons induce HIV expression in latently infected microglial cells. After 48-72 h co-culture, low levels of HIV expression appear to damage neurons, which further enhances HIV expression. There was a marked reduction in intact dendrites staining for microtubule associated protein 2 (MAP2) in the neurons exposed to HIV-expressing microglial cells, indicating extensive dendritic pruning. To model neurotoxicity induced by methamphetamine (METH), we treated cells with nM levels of METH and suboptimal levels of poly (I:C), a TLR3 agonist that mimics the effects of the circulating bacterial rRNA found in HIV infected patients. This combination of agents potently induced HIV expression, with the METH effect mediated by the σ1 receptor (σ1R). In co-cultures of HC69 cells with iCort neurons, the combination of METH and poly(I:C) induced HIV expression and dendritic damage beyond levels seen using either agent alone, Thus, our results demonstrate that the cross-talk between healthy neurons and microglia modulates HIV expression, while HIV expression impairs this intrinsic molecular mechanism resulting in the excessive and uncontrolled stimulation of microglia-mediated neurotoxicity.
Collapse
Affiliation(s)
- David Alvarez-Carbonell
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Fengchun Ye
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Nirmala Ramanath
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Yoelvis Garcia-Mesa
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Pamela E. Knapp
- Departments of Pharmacology and Toxicology and Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States of America
| | - Kurt F. Hauser
- Departments of Pharmacology and Toxicology and Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States of America
| | - Jonathan Karn
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, Ohio, United States of America
| |
Collapse
|
12
|
Lundy DJ, Lee KJ, Peng IC, Hsu CH, Lin JH, Chen KH, Tien YW, Hsieh PCH. Inducing a Transient Increase in Blood-Brain Barrier Permeability for Improved Liposomal Drug Therapy of Glioblastoma Multiforme. ACS NANO 2019; 13:97-113. [PMID: 30532951 DOI: 10.1021/acsnano.8b03785] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The blood-brain barrier (BBB) selectively controls the passage of endogenous and exogenous molecules between systemic circulation and the brain parenchyma. Nanocarrier-based drugs such as liposomes and nanoparticles are an attractive prospect for cancer therapy since they can carry a drug payload and be modified to improve targeting and retention at the desired site. However, the BBB prevents most therapeutic drugs from entering the brain, including physically restricting the passage of liposomes and nanoparticles. In this paper, we show that a low dose of systemically injected recombinant human vascular endothelial growth factor induces a short period of increased BBB permeability. We have shown increased delivery of a range of nanomedicines to the brain including contrast agents for imaging, varying sizes of nanoparticles, small molecule chemotherapeutics, tracer dyes, and liposomal chemotherapeutics. However, this effect was not uniform across all brain regions, and permeability varied depending on the drug or molecule measured. We have found that this window of BBB permeability effect is transient, with normal BBB integrity restored within 4 h. This strategy, combined with liposomal doxorubicin, was able to significantly extend survival in a mouse model of human glioblastoma. We have found no evidence of systemic toxicity, and the technique was replicated in pigs, demonstrating that this technique could be scaled up and potentially be translated to the clinic, thus allowing the use of nanocarrier-based therapies for brain disorders.
Collapse
Affiliation(s)
- David J Lundy
- Institute of Biomedical Sciences , Academia Sinica , Taipei 115 , Taiwan
- Graduate Institute of Biomedical Materials and Tissue Engineering , Taipei Medical University , Taipei 110 , Taiwan
| | - Keng-Jung Lee
- Institute of Biomedical Sciences , Academia Sinica , Taipei 115 , Taiwan
| | - I-Chia Peng
- Institute of Biomedical Sciences , Academia Sinica , Taipei 115 , Taiwan
| | - Chia-Hsin Hsu
- Institute of Biomedical Sciences , Academia Sinica , Taipei 115 , Taiwan
| | - Jen-Hao Lin
- Institute of Biomedical Sciences , Academia Sinica , Taipei 115 , Taiwan
| | - Kun-Hung Chen
- Institute of Biomedical Sciences , Academia Sinica , Taipei 115 , Taiwan
| | - Yu-Wen Tien
- Department of Surgery , National Taiwan University and Hospital , Taipei 100 , Taiwan
| | - Patrick C H Hsieh
- Institute of Biomedical Sciences , Academia Sinica , Taipei 115 , Taiwan
- Department of Surgery , National Taiwan University and Hospital , Taipei 100 , Taiwan
- Institute of Medical Genomics and Proteomics , National Taiwan University , Taipei 100 , Taiwan
- Institute of Clinical Medicine , National Taiwan University , Taipei 100 , Taiwan
| |
Collapse
|
13
|
Roqué PJ, Costa LG. Co-Culture of Neurons and Microglia. CURRENT PROTOCOLS IN TOXICOLOGY 2017; 74:11.24.1-11.24.17. [PMID: 29117434 PMCID: PMC5774987 DOI: 10.1002/cptx.32] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Microglia, the resident immune cells of the brain, have been implicated in numerous neurodegenerative and neurodevelopmental diseases. Activation of microglia by a variety of stimuli induces the release of factors, including pro- and anti-inflammatory cytokines and reactive oxygen species, that contribute to modulating neuro-inflammation and oxidative stress, two crucial processes linked to disorders of the central nervous system. The in vitro techniques described here will provide a set of protocols for the isolation and plating of primary cerebellar granule neurons, primary cortical microglia from a mixed glia culture, and methods for co-culturing both cell types. These methods allow the study of how microglia and the factors they release in this shared environment mediate the effects of toxicants on neuronal function and survival. The protocols presented here allow for flexibility in experimental design, the study of numerous toxicological endpoints, and the opportunity to explore neuroprotective strategies. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
| | - Lucio G. Costa
- University of Washington, Seattle, WA
- University of Parma, Parma, Italy
| |
Collapse
|
14
|
Kuhlman KR, Chiang JJ, Horn S, Bower JE. Developmental psychoneuroendocrine and psychoneuroimmune pathways from childhood adversity to disease. Neurosci Biobehav Rev 2017; 80:166-184. [PMID: 28577879 PMCID: PMC5705276 DOI: 10.1016/j.neubiorev.2017.05.020] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 04/13/2017] [Accepted: 05/22/2017] [Indexed: 12/17/2022]
Abstract
Childhood adversity has been repeatedly and robustly linked to physical and mental illness across the lifespan. Yet, the biological pathways through which this occurs remain unclear. Functioning of the inflammatory arm of the immune system and the hypothalamic-pituitary-adrenal (HPA)-axis are both hypothesized pathways through which childhood adversity leads to disease. This review provides a novel developmental framework for examining the role of adversity type and timing in inflammatory and HPA-axis functioning. In particular, we identify elements of childhood adversity that are salient to the developing organism: physical threat, disrupted caregiving, and unpredictable environmental conditions. We propose that existing, well-characterized animal models may be useful in differentiating the effects of these adversity elements and review both the animal and human literature that supports these ideas. To support these hypotheses, we also provide a detailed description of the development and structure of both the HPA-axis and the inflammatory arm of the immune system, as well as recent methodological advances in their measurement. Recommendations for future basic, developmental, translational, and clinical research are discussed.
Collapse
|
15
|
Humbert-Claude M, Duc D, Dwir D, Thieren L, Sandström von Tobel J, Begka C, Legueux F, Velin D, Maillard MH, Do KQ, Monnet-Tschudi F, Tenenbaum L. Tollip, an early regulator of the acute inflammatory response in the substantia nigra. J Neuroinflammation 2016; 13:303. [PMID: 27927222 PMCID: PMC5142340 DOI: 10.1186/s12974-016-0766-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 11/18/2016] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Tollip is a ubiquitously expressed protein, originally described as a modulator of the IL-1R/TLR-NF-κB signaling pathways. Although this property has been well characterized in peripheral cells, and despite some evidence of its expression in the central nervous system, the role of Tollip in neuroinflammation remains poorly understood. The present study sought to explore the implication of Tollip in inflammation in the substantia nigra pars compacta, the structure affected in Parkinson's disease. METHODS We first investigated Tollip distribution in the midbrain by immunohistochemistry. Then, we addressed TLR4-mediated response by intra-nigral injections of lipopolysaccharide (LPS), a TLR4 agonist, on inflammatory markers in Tollip knockout (KO) and wild-type (WT) mice. RESULTS We report an unexpectedly high Tollip immunostaining in dopaminergic neurons of the mice brain. Second, intra-nigral injection of LPS led to increased susceptibility to neuroinflammation in Tollip KO compared to Tollip WT mice. This was demonstrated by a significant increase of tumor necrosis factor alpha (TNF-α), interleukin 1 beta (IL-1β), interleukin 6 (IL-6), and interferon gamma (IFN-γ) messenger RNA (mRNA) in the midbrain of Tollip KO mice upon LPS injection. Consistently, brain rAAV viral vector transduction with a nuclear factor kappa B (NF-κB)-inducible reporter gene confirmed increased NF-κB activation in Tollip KO mice. Lastly, Tollip KO mice displayed higher inducible NO synthase (iNOS) production, both at the messenger and protein level when compared to LPS-injected WT mice. Tollip deletion also aggravated LPS-induced oxidative and nitrosative damages, as indicated by an increase of 8-oxo-2'-deoxyguanosine and nitrotyrosine immunostaining, respectively. CONCLUSIONS Altogether, these findings highlight a critical role of Tollip in the early phase of TLR4-mediated neuroinflammation. As brain inflammation is known to contribute to Parkinson's disease, Tollip may be a potential target for neuroprotection.
Collapse
Affiliation(s)
- Marie Humbert-Claude
- Laboratory of Cellular and Molecular Neurotherapies, Center for Neuroscience Research, Department of Clinical Neuroscience, Lausanne University Hospital, Lausanne, Switzerland
| | - D. Duc
- Laboratory of Cellular and Molecular Neurotherapies, Center for Neuroscience Research, Department of Clinical Neuroscience, Lausanne University Hospital, Lausanne, Switzerland
| | - D. Dwir
- Department of Psychiatry, Center for Psychiatric Neuroscience, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Prilly, Lausanne, Switzerland
| | - L. Thieren
- Laboratory of Cellular and Molecular Neurotherapies, Center for Neuroscience Research, Department of Clinical Neuroscience, Lausanne University Hospital, Lausanne, Switzerland
| | | | - C. Begka
- Service of Gastroenterology and Hepatology, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | | | - D. Velin
- Service of Gastroenterology and Hepatology, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - M. H. Maillard
- Service of Gastroenterology and Hepatology, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - K. Q. Do
- Department of Psychiatry, Center for Psychiatric Neuroscience, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Prilly, Lausanne, Switzerland
| | - F. Monnet-Tschudi
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - L. Tenenbaum
- Laboratory of Cellular and Molecular Neurotherapies, Center for Neuroscience Research, Department of Clinical Neuroscience, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
16
|
Kern JK, Geier DA, Sykes LK, Geier MR. Relevance of Neuroinflammation and Encephalitis in Autism. Front Cell Neurosci 2016; 9:519. [PMID: 26834565 PMCID: PMC4717322 DOI: 10.3389/fncel.2015.00519] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Accepted: 12/24/2015] [Indexed: 12/12/2022] Open
Abstract
In recent years, many studies indicate that children with an autism spectrum disorder (ASD) diagnosis have brain pathology suggestive of ongoing neuroinflammation or encephalitis in different regions of their brains. Evidence of neuroinflammation or encephalitis in ASD includes: microglial and astrocytic activation, a unique and elevated proinflammatory profile of cytokines, and aberrant expression of nuclear factor kappa-light-chain-enhancer of activated B cells. A conservative estimate based on the research suggests that at least 69% of individuals with an ASD diagnosis have microglial activation or neuroinflammation. Encephalitis, which is defined as inflammation of the brain, is medical diagnosis code G04.90 in the International Classification of Disease, 10th revision; however, children with an ASD diagnosis are not generally assessed for a possible medical diagnosis of encephalitis. This is unfortunate because if a child with ASD has neuroinflammation, then treating the underlying brain inflammation could lead to improved outcomes. The purpose of this review of the literature is to examine the evidence of neuroinflammation/encephalitis in those with an ASD diagnosis and to address how a medical diagnosis of encephalitis, when appropriate, could benefit these children by driving more immediate and targeted treatments.
Collapse
Affiliation(s)
- Janet K. Kern
- Institute of Chronic Illnesses, Inc., Silver SpringMD, USA
| | - David A. Geier
- Institute of Chronic Illnesses, Inc., Silver SpringMD, USA
| | | | - Mark R. Geier
- Institute of Chronic Illnesses, Inc., Silver SpringMD, USA
| |
Collapse
|
17
|
Direct angiotensin type 2 receptor (AT2R) stimulation attenuates T-cell and microglia activation and prevents demyelination in experimental autoimmune encephalomyelitis in mice. Clin Sci (Lond) 2014; 128:95-109. [PMID: 25052203 DOI: 10.1042/cs20130601] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In the present study, we evaluated stimulation of the angiotensin type 2 receptor (AT2R) by the selective non-peptide agonist Compound 21 (C21) as a novel therapeutic concept for the treatment of multiple sclerosis using the model of experimental autoimmune encephalomyelitis (EAE) in mice. C57BL-6 mice were immunized with myelin-oligodendrocyte peptide and treated for 4 weeks with C21 (0.3 mg/kg/day i.p.). Potential effects on myelination, microglia and T-cell composition were estimated by immunostaining and FACS analyses of lumbar spinal cords. The in vivo study was complemented by experiments in aggregating brain cell cultures and microglia in vitro. In the EAE model, treatment with C21 ameliorated microglia activation and decreased the number of total T-cells and CD4+ T-cells in the spinal cord. Fluorescent myelin staining of spinal cords further revealed a significant reduction in EAE-induced demyelinated areas in lumbar spinal cord tissue after AT2R stimulation. C21-treated mice had a significantly better neurological score than vehicle-treated controls. In aggregating brain cell cultures challenged with lipopolysaccharide (LPS) plus interferon-γ (IFNγ), AT2R stimulation prevented demyelination, accelerated re-myelination and reduced the number of microglia. Cytokine synthesis and nitric oxide production by microglia in vitro were significantly reduced after C21 treatment. These results suggest that AT2R stimulation protects the myelin sheaths in autoimmune central nervous system inflammation by inhibiting the T-cell response and microglia activation. Our findings identify the AT2R as a potential new pharmacological target for demyelinating diseases such as multiple sclerosis.
Collapse
|
18
|
Noriega DB, Savelkoul HFJ. Immune dysregulation in autism spectrum disorder. Eur J Pediatr 2014; 173:33-43. [PMID: 24297668 DOI: 10.1007/s00431-013-2183-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 10/09/2013] [Indexed: 12/25/2022]
Abstract
UNLABELLED Autism spectrum disorder (ASD) is a common and severe neuro-developmental disorder in early childhood which is defined by social and communication deficits and repetitive and stereotypic behaviours. The aetiology of ASD remains poorly understood. Susceptibility to development of ASD has significant environmental components, in addition to the profound genetic heritability. Few genes have been associated to the risk for ASD development. There is substantial evidence implicating chronic neurological inflammation and immune dysregulation leading to upregulation of inflammatory cytokines in the ASD brain, probably due to altered blood-brain barrier function. The immune system is characterized by excessive and skewed cytokine responses, modulated T cell reactivity, decreased regulation and production of immunosuppressive cytokines, modified NK function and increased autoantibody production. CONCLUSION The perinatal environment generates vulnerability to chronic neuro-inflammation in the brain associated with profound modulation and dysregulation in the immune system leading to the rapid development of ASD in genetically susceptible children.
Collapse
Affiliation(s)
- Daniela Briceno Noriega
- Cell Biology and Immunology Group, Wageningen University, P.O. Box 338, 6700 AH, Wageningen, The Netherlands
| | | |
Collapse
|
19
|
Kempuraj D, Khan MM, Thangavel R, Xiong Z, Yang E, Zaheer A. Glia maturation factor induces interleukin-33 release from astrocytes: implications for neurodegenerative diseases. J Neuroimmune Pharmacol 2013; 8:643-50. [PMID: 23397250 DOI: 10.1007/s11481-013-9439-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 01/28/2013] [Indexed: 01/09/2023]
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD) and Multiple sclerosis (MS) involve activation of glial cells and release of inflammatory mediators leading to death of neurons. Glia maturation factor (GMF) is up-regulated in the central nervous system (CNS) in these neurodegenerative diseases. Interleukin-33 (IL-33) is highly expressed constitutively in the CNS. We have treated mouse astrocytes, mixed culture with glial cells and neurons, and only neurons with GMF and/or IL-33 in vitro. Both GMF and IL-33-induced chemokine (C-C motif) ligand 2 (CCL2) release in a dose and time-dependent manner. We report that GMF induced IL-33 release, and that IL-33 augments GMF-induced tumor necrosis factor-alpha (TNF-α) release from mouse astrocytes. IL-33 induces CCL2, TNF-α and nitric oxide release through phosphorylation of ERK in mouse astrocytes. Incubation of mixed culture containing glial cells and neurons or only neuronal culture with IL-33 reduced the number of neurons positive for microtubule-associated protein 2. In conclusion, IL-33 augments GMF-mediated neuroinflammation and may provide a new drug target for neurodegenerative and autoimmune diseases.
Collapse
|
20
|
Evidence of microglial activation in autism and its possible role in brain underconnectivity. ACTA ACUST UNITED AC 2012; 7:205-13. [PMID: 22874006 PMCID: PMC3523548 DOI: 10.1017/s1740925x12000142] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Evidence indicates that children with autism spectrum disorder (ASD) suffer from an
ongoing neuroinflammatory process in different regions of the brain involving microglial
activation. When microglia remain activated for an extended period, the production of
mediators is sustained longer than usual and this increase in mediators contributes to
loss of synaptic connections and neuronal cell death. Microglial activation can then
result in a loss of connections or underconnectivity. Underconnectivity is reported in
many studies in autism. One way to control neuroinflammation is to reduce or inhibit
microglial activation. It is plausible that by reducing brain inflammation and microglial
activation, the neurodestructive effects of chronic inflammation could be reduced and
allow for improved developmental outcomes. Future studies that examine treatments that may
reduce microglial activation and neuroinflammation, and ultimately help to mitigate
symptoms in ASD, are warranted.
Collapse
|