1
|
Sehgal N, Li L, Goin DE, Chen J, Jigmeddagva U, Morello-Frosch R, Woodruff TJ, Gaw SL, Robinson JF, Eick SM. Psychosocial stress and associations with inflammation in mid-gestation maternal, fetal, and placental tissue. Reprod Toxicol 2025; 135:108922. [PMID: 40254104 DOI: 10.1016/j.reprotox.2025.108922] [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: 01/28/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND Inflammation has been implicated as an intermediary between psychosocial stress and adverse birth outcomes. However, prior work has mostly relied on maternal inflammation as a proxy for fetal inflammation mid-gestation or measured fetal inflammation in cord blood and placenta obtained at delivery. No studies have examined psychosocial stress in relation to fetal inflammation mid-gestation. METHODS Twenty cytokines were measured in matched maternal blood, cord blood, and placenta obtained mid-gestation from a socio-demographically diverse group of pregnant participants undergoing elective second-trimester pregnancy terminations (N = 106). Corticotropin-releasing hormone, a proposed biomarker of gestational length, was measured in maternal blood. Perceived stress, and exposure to stressful life events, job strain, and social support were measured via questionnaires. We used linear regression to estimate associations between individual stressors and inflammatory biomarkers in each biomatrix and principal component analysis to assess groups of inflammatory biomarkers. RESULTS We observed many matrix-specific associations between psychosocial stressors and inflammatory biomarkers. For example, low versus high social support was associated with significantly decreased levels of maternal blood CCL3 (β=-0.53; 95 % confidence interval [CI]=-0.98,-0.07), CCL4 (β=-0.26; 95 % CI=-0.47,-0.04), IL8 (β=-0.79; 95 % CI=-1.47,-0.11), CXCL9 (β=-0.47; 95 % CI=-0.89,-0.06), IFNγ (β=-2.28; 95 % CI=-3.60,-0.96), IL4 (β=-1.07; 95 % CI=-1.88,-0.26); and cord blood IFNγ (β=-0.83; 95 % CI=-1.52,-0.14). Social support was not associated with placental inflammation. CONCLUSIONS During mid-pregnancy, psychosocial stress─ particularly low social support─ was associated with maternal blood levels of select cytokines, suggesting a potential pathway linking social stress and inflammation. Our results indicate that the placenta may buffer these inflammatory effects on the fetus.
Collapse
Affiliation(s)
- Neha Sehgal
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Lin Li
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Dana E Goin
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Jessica Chen
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Unurzul Jigmeddagva
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Rachel Morello-Frosch
- Program on Reproductive Health and the Environment, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA; Department of Environmental Science, Policy and Management and School of Public Health, University of California, Berkeley, CA, USA
| | - Tracey J Woodruff
- Program on Reproductive Health and the Environment, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Stephanie L Gaw
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA; Division of Maternal Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Joshua F Robinson
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA; Program on Reproductive Health and the Environment, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Stephanie M Eick
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA.
| |
Collapse
|
2
|
AlSharari SD, Mahmood HM, Alasmari AF, AlDhalaan HM, Alasmari F, Khan MR, Ahmad SF, Aljasham AT, Damaj IM, Alshammari MA. Nicotine Attenuates Molecular Signalings in the BTBR T + Itpr3 tf/J Mouse Model of Autism. Mol Neurobiol 2025:10.1007/s12035-025-04894-6. [PMID: 40172818 DOI: 10.1007/s12035-025-04894-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 03/24/2025] [Indexed: 04/04/2025]
Abstract
Accumulating evidence indicates that nicotinic receptor subtypes are altered in the brains of autistic individuals, and nicotinic acetylcholine receptors (nAChRs) play essential roles in autistic profiles in BTBR T+ Itpr3tf/J mice. This study aimed to elucidate the roles of nicotine on systemic inflammatory cytokine levels and expression patterns of nicotinic receptor subtypes in the prefrontal cortex in BTBR T+ Itpr3tf/J mice. This research project characterized the effect of chronic treatment with nicotine at a dose (100 mcg/ml; po) administrated orally in drinking water over a period of fourteen days in BTBR T+ Itpr3tf/J mice, while C57BL/6 J mice were served as the controls. Following the nicotine treatment, the levels of tumor necrosis factor (TNF)-α, interferon (IFN)-γ interleukin (IL)-1β, and granulocyte-macrophage colony-stimulating factor (GM-CSF) were assessed in the serum; the levels of pro-inflammatory cytokines [interleukin (IL)-17 and interferon (IFN)-γ], on CD4+ and CD8+ T cells were evaluated in the blood. Moreover, the expression of α7, α4, and β2-nAChRs in the prefrontal cortex in BTBR T+ Itpr3tf/J mice was examined. Biochemical analysis showed that nicotine had significantly decreased the concentration of inflammatory cytokines, including TNF-α, IFN-γ, IL-1β, and GM-CSF in the serum, and reduced the expression levels of intracellular pro-inflammatory cytokines (IL-17 & IFN-γ) on CD4+ and CD8+ T cells in the blood while mecamylamine reversed the effect of IL-17+ CD4+ T cells. Nicotine administration up-regulated the expressions of α7, α4, and β2 nAChRs in the prefrontal cortex in BTBR T+ Itpr3tf/J mice. The current results indicate that nAChRs play a significant role, at least in part, in ASD and might serve as a crucial target for therapeutic interventions in ASD.
Collapse
Affiliation(s)
- Shakir D AlSharari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| | - Hafiz M Mahmood
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah F Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hesham M AlDhalaan
- Department of Neuroscience, Center for Autism Research, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad R Khan
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Alanoud T Aljasham
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Imad M Damaj
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Musaad A Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
3
|
Mortamais M, Ongono JS, Michelon C, Hough I, Seyve E, Kloog I, Zaros C, Charles MA, Lepeule J, Baghdadli A. Prenatal Exposure to Ambient Particulate Matter and Autism Spectrum Disorder in Children, a Case Control Study in France. J Autism Dev Disord 2025:10.1007/s10803-025-06795-y. [PMID: 40120015 DOI: 10.1007/s10803-025-06795-y] [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] [Accepted: 03/03/2025] [Indexed: 03/25/2025]
Abstract
A series of epidemiological studies conducted in the United States have consistently shown an increased risk of autism spectrum disorder (ASD) in children associated with pre- and postnatal exposure to ambient particulate matter (PM). In Europe, studies are scarce and results are inconsistent. We aimed to investigate the association between prenatal exposure to PM and the risk of ASD in France. ASD cases were participants from the ELENA cohort. Controls children from the ELFE cohort were matched by sex, year (± 2) and region of birth. Prenatal exposures to PM10 and to PM2.5 were estimated between 2008 and 2013 using innovative hybrid spatio-temporal models developed for France. Conditional logistic regression models adjusted for birth season, parent's age at the child birth and parental education level were run. We included 125 ASD cases and 500 controls. Prenatal PM2.5 and PM10 median (IQR) concentration estimates were respectively 16.3 (3.9) µg/m3 and 22.9 (6.6) µg/m3 in the whole sample. The conditional logistic regression models showed Odds Ratios (ORs) (Confidence Interval 95%) for ASD risk of 0.72 (0.52-1.01) and 0.84 (0.58-1.22) for an IQR increase in PM2.5 and PM10 prenatal levels, respectively. When restricting population of ASD cases to children born the same year of controls, ORs were 1.79 (0.80-4.01) and 2.23 (0.71-9.04), respectively. Our results did not show that prenatal exposures to PM2.5 and PM10 were associated with the risk of ASD in children in France.Trial Registration Number NCT02625116.
Collapse
Affiliation(s)
- Marion Mortamais
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, Inserm, Montpellier, France
| | - Jeanne Sandrine Ongono
- Centre de Ressource Autisme Languedoc-Roussillon et Centre d'Excellence sur l'Autisme et les Troubles Neuro-développementaux (CeAND), 291 Avenue du Doyen Giraud, Montpellier, 34000, France
| | - Cécile Michelon
- Centre de Ressource Autisme Languedoc-Roussillon et Centre d'Excellence sur l'Autisme et les Troubles Neuro-développementaux (CeAND), 291 Avenue du Doyen Giraud, Montpellier, 34000, France
| | - Ian Hough
- University Grenoble Alpes, Inserm, CNRS, Team of Environmental Epidemiology Applied to Development and Respiratory Health, IAB, Grenoble, France
| | - Emie Seyve
- University Grenoble Alpes, Inserm, CNRS, Team of Environmental Epidemiology Applied to Development and Respiratory Health, IAB, Grenoble, France
- France & Université de Paris Cité, Inserm, INRAE, Center of Research in Epidemiology and StatisticS (CRESS), Paris, 75004, France
| | - Itai Kloog
- Department of Geography and Environmental Development, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Cécile Zaros
- French Institute for Demographic Studies (Ined), French Institute for Medical Research and Health (Inserm), French Blood Agency, ELFE Joint Unit, Paris, France
| | - Marie-Aline Charles
- French Institute for Demographic Studies (Ined), French Institute for Medical Research and Health (Inserm), French Blood Agency, ELFE Joint Unit, Paris, France
- French Institute for Medical Research and Health, UMR 1153 Centre for Research in Epidemiology and Statistics Paris University (CRESS), Team Early Life Research on Later Health, Villejuif, France
| | - Johanna Lepeule
- University Grenoble Alpes, Inserm, CNRS, Team of Environmental Epidemiology Applied to Development and Respiratory Health, IAB, Grenoble, France
| | - Amaria Baghdadli
- Centre de Ressource Autisme Languedoc-Roussillon et Centre d'Excellence sur l'Autisme et les Troubles Neuro-développementaux (CeAND), 291 Avenue du Doyen Giraud, Montpellier, 34000, France.
- Université Paris-Saclay, UVSQ, Inserm, CESP, Team DevPsy, Villejuif, 94807, France.
| |
Collapse
|
4
|
Sidhu RK, Maparu K, Singh S, Aran KR. Unveiling the role of Na⁺/K⁺-ATPase pump: neurodegenerative mechanisms and therapeutic horizons. Pharmacol Rep 2025:10.1007/s43440-025-00717-6. [PMID: 40117043 DOI: 10.1007/s43440-025-00717-6] [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: 12/27/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/23/2025]
Abstract
Sodium and potassium-activated adenosine 5'-triphosphatase (Na+/K+-ATPase) is a pivotal plasma membrane enzyme involved in neuronal activity and cellular homeostasis. The dysregulation of these enzymes has been implicated in a spectrum of neurodegenerative disorders like Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and neurodevelopmental disorders including autism spectrum disorder (ASD), psychiatric disorders such as schizophrenia, and neurological problems like epilepsy. A hallmark of these disorders is the gradual loss of neuronal integrity and function, often exacerbated by protein accumulation within brain cells. This review delves into the multifaceted role of Na+/K+-ATPase dysfunction in driving oxidative stress, excitotoxicity, and neuroinflammation, contributing to synaptic and neuronal damage. Emerging therapeutic strategies, such as gene therapy and developing isoform-specific enzyme modulators, offer promising avenues for targeted interventions. Furthermore, this review highlights innovative research directions, including the role of Na⁺/K⁺-ATPase in synaptic plasticity, the identification of endogenous regulators, and its contribution to neuroinflammatory pathways. Personalized medicine and advanced gene-editing technologies are positioned as transformative tools for crafting safer and more precise therapies tailored to individual patients. This comprehensive exploration underscores the enzyme's therapeutic potential and sets the stage for developing novel targeted strategies to mitigate the burden of Na⁺/K⁺-ATPase-linked neurological disorders.
Collapse
Affiliation(s)
- Ramandeep Kaur Sidhu
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Kousik Maparu
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Shamsher Singh
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Khadga Raj Aran
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| |
Collapse
|
5
|
Davis LK, Ince LM, Gullapalli S, Fonken LK. Neuroimmune and behavioral changes elicited by maternal immune activation in mice are ameliorated by early postnatal immune stimulation. Brain Behav Immun 2025; 127:375-386. [PMID: 40081778 DOI: 10.1016/j.bbi.2025.03.005] [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: 09/17/2024] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025] Open
Abstract
Though the etiology of autism spectrum disorder (ASD) is complex and not fully understood, it is believed that genetic risk factors, coupled with early life inflammation may predispose individuals to develop ASD. Maternal immune activation (MIA) is associated with increased incidence of ASD in offspring; however, not all mothers who experience inflammation during pregnancy have children with autism, suggesting that MIA may act as a disease primer that results in ASD pathology when paired with additional inflammatory insults. Here, we tested the hypothesis that MIA is a disease primer by using a two-hit model that combined MIA with a secondary immune stimulation in early life. C57BL/6J mouse dams were treated with polyinosinic-polycytidylic acid (Poly(I:C)) at embyronic day 12.5, and a subset of litters were then treated with the endotoxin lipopolysaccharide (LPS) four days after birth. Offspring were assessed in young adulthood for changes in behavior including sociability, repetitive-like behaviors, and anxiety-like behaviors. Flow cytometry was performed in adulthood to assess changes in immune cell populations in the periphery and in the brain. MIA increased repetitive-like behaviors in male mice and decreased sociability in both sexes. Unexpectedly, the secondary immune stimulation with LPS did not exacerbate changes in social and repetitive-like behaviors in either sex. MIA also altered distribution of cytotoxic CD8 + T cell populations in the periphery and brain of both sexes: CD8 + T cells were elevated in thymus but reduced in spleen, lymph, and brain. Additionally, MIA altered microglia activity in a region-specific manner in male mice, which was also not exacerbated but rather ameliorated when combined with LPS. Our results demonstrate that changes in repetitive-like and social behaviors that are induced by MIA in male mice are not exacerbated by subsequent inflammatory challenge and highlights the importance of considering the timing of stressors in the appearance of developmental pathology.
Collapse
Affiliation(s)
- Lourdes K Davis
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA; Interdisciplinary Neuroscience Program, University of Texas at Austin, Austin, TX 78712, USA.
| | - Louise M Ince
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Sriya Gullapalli
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Laura K Fonken
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA; Interdisciplinary Neuroscience Program, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
6
|
Hushmandi K, Reiter RJ, Farahani N, Cho WC, Alimohammadi M, Khoshnazar SM. Pyroptosis; igniting neuropsychiatric disorders from mild depression to aging-related neurodegeneration. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111325. [PMID: 40081561 DOI: 10.1016/j.pnpbp.2025.111325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Neuropsychiatric disorders significantly impact global health and socioeconomic well-being, highlighting the urgent need for effective treatments. Chronic inflammation, often driven by the innate immune system, is a key feature of many neuropsychiatric conditions. NOD-like receptors (NLRs), which are intracellular sensors, detect danger signals and trigger inflammation. Among these, NLR protein (NLRP) inflammasomes play a crucial role by releasing pro-inflammatory cytokines and inducing a particular cell death process known as pyroptosis. Pyroptosis is defined as a proinflammatory form of programmed cell death executed by cysteine-aspartic proteases, also known as caspases. Currently, the role of pyroptotic flux has emerged as a critical factor in innate immunity and the pathogenesis of multiple diseases. Emerging evidence suggests that the induction of pyroptosis, primarily due to NLRP inflammasome activation, is involved in the pathophysiology of various neuropsychiatric disorders, including depression, stress-related issues, schizophrenia, autism spectrum disorders, and neurodegenerative diseases. Within this framework, the current review explores the complex relationship between pyroptosis and neuropsychiatric diseases, aiming to identify potential therapeutic targets for these challenging conditions.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, TX, USA
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyedeh Mahdieh Khoshnazar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
7
|
Díaz-Pons A, Castaño-Castaño S, Ortiz-García de la Foz V, Yorca-Ruiz Á, Martínez-Asensi C, Munarriz-Cuezva E, Ayesa-Arriola R. Understanding the potential impact of trimester-specific maternal immune activation due to SARS-CoV-2 on early human neurodevelopment and the role of cytokine balance. Brain Behav Immun Health 2025; 44:100956. [PMID: 39990281 PMCID: PMC11846590 DOI: 10.1016/j.bbih.2025.100956] [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: 09/30/2024] [Revised: 12/23/2024] [Accepted: 01/22/2025] [Indexed: 02/25/2025] Open
Abstract
Purpose The COVID-19 pandemic presents significant future health challenges. Its impact on pregnant women and their newborn is a particular area of concern. This study aims to examine the potential role of maternal immune activation (MIA), due to SARS-CoV-2 infection, on early neurodevelopment. Methods We analysed 107 mother-infant dyads from the COGESTCOV-19 study in Cantabria, Spain, which included 59 SARS-CoV-2 exposed (cases) and 48 unexposed (controls) mothers, recruited between December 2020 and February 2022. Cytokine levels (IL-6 and IL-10) were obtained from maternal blood and cord blood. Neurodevelopment was assessed using the Neonatal Behavioral Assessment Scale (NBAS) at six weeks of age. Trimester of infection was considered in the main analyses. Results Results showed no significant overall delays in early neurodevelopment due to maternal SARS-CoV-2 infection. Control infants performed better in some NBAS items. However, cases infants showed trimester-specific differences. First-trimester exposure was related to motor and reflex delays, second-trimester to poorer performances in motor tasks and autonomic stability, and third-trimester to weaker state organization, regulation, and reflexes. Some correlations between cytokine levels and NBAS performance showed moderate associations. Conclusions These findings highlight the need for ongoing neurodevelopmental monitoring of infants born during the COVID-19 pandemic. The study enhances our understanding of MIA's impact on early development, emphasizing the importance of addressing homeostatic mechanisms in mothers and newborns.
Collapse
Affiliation(s)
- Alexandre Díaz-Pons
- Departamento de Investigación en Enfermedades Mentales, Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain
- Escuela de Doctorado de la Universidad de Cantabria (EDUC), Universidad de Cantabria (UC), 39005, Santander, Spain
- Departamento de Medicina y Ciencias de la Salud, Facultad de Medicina, Universidad de Cantabria (UC), 39011, Santander, Spain
- Facultad de Psicología, Universidad Nacional de Educación a Distancia (UNED), 28015, Madrid, Spain
| | - Sergio Castaño-Castaño
- Departamento de Psicobiología, Facultad de Psicología, Universidad de Oviedo (UO), 33003, Oviedo, Spain
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33003, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011, Oviedo, Spain
| | - Víctor Ortiz-García de la Foz
- Departamento de Investigación en Enfermedades Mentales, Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain
| | - Ángel Yorca-Ruiz
- Departamento de Investigación en Enfermedades Mentales, Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain
- Escuela de Doctorado de la Universidad de Cantabria (EDUC), Universidad de Cantabria (UC), 39005, Santander, Spain
- Departamento de Biología Molecular, Facultad de Medicina, Universidad de Cantabria (UC), 39011, Santander, Spain
| | - Carlos Martínez-Asensi
- Departamento de Investigación en Enfermedades Mentales, Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain
- Facultad de Psicología, Universidad Nacional de Educación a Distancia (UNED), 28015, Madrid, Spain
| | - Eva Munarriz-Cuezva
- Departamento de Farmacología, Facultad de Medicina y Enfermería, Universidad del País Vasco/ Euskal Herriko Unibertsitatea (UPV/EHU), 48940, Leioa, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Rosa Ayesa-Arriola
- Departamento de Investigación en Enfermedades Mentales, Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain
- Departamento de Medicina y Ciencias de la Salud, Facultad de Medicina, Universidad de Cantabria (UC), 39011, Santander, Spain
- Facultad de Psicología, Universidad Nacional de Educación a Distancia (UNED), 28015, Madrid, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, 28029, Madrid, Spain
| |
Collapse
|
8
|
O'Grady K, Grabrucker AM. Metal Dyshomeostasis as a Driver of Gut Pathology in Autism Spectrum Disorders. J Neurochem 2025; 169:e70041. [PMID: 40108935 PMCID: PMC11923526 DOI: 10.1111/jnc.70041] [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: 10/01/2024] [Revised: 01/31/2025] [Accepted: 02/25/2025] [Indexed: 03/22/2025]
Abstract
Despite being classified as neurodevelopmental disorders, in recent years, there has been a growing interest in the association between autism spectrum disorders (ASDs) and gut pathology. This comprehensive and systematic review explores a potential mechanism underlying gut pathology in ASDs, including alterations in gut microbiota, intestinal permeability, immune dysregulation, and gastrointestinal (GI) symptoms. Specifically, it delves into the role of toxic and essential metals and their interplay, affecting the development and function of the GI tract. The review also discusses the potential implications of this gut pathology in the development and management of ASDs. Studies have shown that heavy metal exposure, whether through environmental sources or dietary intake, can disrupt the delicate balance of trace elements in the gut. This disruption can adversely affect zinc homeostasis, potentially exacerbating gut pathology in individuals with ASDs. The impaired zinc absorption resulting from heavy metal exposure may contribute to the immune dysregulation, oxidative stress, and inflammation observed in the gut of individuals with ASDs. By shedding light on the multifaceted nature of gut pathology, including the impact of metal dyshomeostasis as a non-genetic factor in ASD, this review underscores the significance of the gut-brain axis in the etiology and management of ASDs.
Collapse
Affiliation(s)
- Katelyn O'Grady
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
- Bernal Institute, University of Limerick, Limerick, Ireland
- Health Research Institute (HRI), University of Limerick, Limerick, Ireland
| | - Andreas M Grabrucker
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
- Bernal Institute, University of Limerick, Limerick, Ireland
- Health Research Institute (HRI), University of Limerick, Limerick, Ireland
| |
Collapse
|
9
|
Campos JMB, de Aguiar da Costa M, de Rezende VL, Costa RRN, Ebs MFP, Behenck JP, de Roch Casagrande L, Venturini LM, Silveira PCL, Réus GZ, Gonçalves CL. Animal Model of Autism Induced by Valproic Acid Combined with Maternal Deprivation: Sex-Specific Effects on Inflammation and Oxidative Stress. Mol Neurobiol 2025; 62:3653-3672. [PMID: 39316355 DOI: 10.1007/s12035-024-04491-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 09/08/2024] [Indexed: 09/25/2024]
Abstract
Autism spectrum disorder (ASD) etiology probably involves a complex interplay of both genetic and environmental risk factors, which includes pre- and perinatal exposure to environmental stressors. Thus, this study evaluated the effects of prenatal exposure to valproic acid (VPA) combined with maternal deprivation (MD) on behavior, oxidative stress parameters, and inflammatory state at a central and systemic level in male and female rats. Pregnant Wistar rats were exposed to VPA during gestation, and the offspring were submitted to MD. Offspring were tested for locomotor and social behavior; rats were euthanized, where the cerebellum, posterior cortex, prefrontal cortex, and peripheric blood were collected for oxidative stress and inflammatory analysis. It was observed that young rats (25-30 days old) exposed only to VPA presented a lower social approach when compared to the control group. VPA + MD rats did not present the same deficit. Female rats exposed to VPA + MD presented oxidative stress in all brain areas analyzed. Male rats in the VPA and VPA + MD groups presented oxidative stress only in the cerebellum. Regarding inflammatory parameters, male rats exposed only to MD exhibited an increase in pro-inflammatory cytokines in the blood and in the cortex total. The same was observed in females exposed only to VPA. Animals exposed to VPA + MD showed no alterations in the cytokines analyzed. In summary, gestational (VPA) and perinatal (MD) insults can affect molecular mechanisms such as oxidative stress and inflammation differently depending on the sex and brain area analyzed. Combined exposition to VPA and MD triggers oxidative stress especially in female brains without evoking an inflammatory response.
Collapse
Affiliation(s)
- José Marcelo Botancin Campos
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), 1105, Criciúma, SC, 88806-000, Brazil
| | - Maiara de Aguiar da Costa
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), 1105, Criciúma, SC, 88806-000, Brazil
| | - Victória Linden de Rezende
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), 1105, Criciúma, SC, 88806-000, Brazil
| | - Rosiane Ronchi Nascimento Costa
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), 1105, Criciúma, SC, 88806-000, Brazil
| | - Maria Fernanda Pedro Ebs
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), 1105, Criciúma, SC, 88806-000, Brazil
| | - João Paulo Behenck
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Laura de Roch Casagrande
- Laboratory of Experimental Physiopathology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Ligia Milanez Venturini
- Laboratory of Experimental Physiopathology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Paulo Cesar Lock Silveira
- Laboratory of Experimental Physiopathology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Gislaine Zilli Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Cinara Ludvig Gonçalves
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), 1105, Criciúma, SC, 88806-000, Brazil.
| |
Collapse
|
10
|
Anindya I, Sekartini R, Ariyanto IA, Wiguna T, Sari NP, Rahayu YS, Soebandrio A. Cytomegalovirus-Reactive IgG Correlates with Increased IL-6 and IL-1β Levels, Affecting Eating Behaviours and Tactile Sensitivity in Children with Autism. Biomedicines 2025; 13:338. [PMID: 40002751 PMCID: PMC11852405 DOI: 10.3390/biomedicines13020338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/06/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Elevated cytokine levels, including IL-6 and IL-1β, can contribute to persistent brain inflammation in children with autism and cytomegalovirus (CMV) infection, exacerbating autism-related behaviours and symptoms. This study evaluates the impact of CMV-induced cytokine increases on the eating behaviours and sensory profiles of children with autism. METHODS A cross-sectional design was employed, involving children aged two to five years (CMV-reactive IgG), with ASD (n= 98) and TD (n = 96). Serological tests using ELISA were conducted to measure IgG CMV, IL-6, and IL-1β biomarkers. Eating behaviours were evaluated using the BAMBI (Brief Autism Mealtime Behaviour Inventory), and sensory profiles were assessed using the SSP (Short Sensory Profile). Statistical analyses were performed using Spearman's rank and chi-square tests. RESULTS The results show that autism significantly affects children's eating behaviours and sensory profiles (p < 0.001), with notable differences found between the groups. Correlation analysis revealed a significant association between IgG CMV and IL-6 (p = 0.026) and IL-1β (p = 0.014) in the ASD group. Additionally, eating behaviours (food refusal and limited variety) in ASD correlated with IL-6 and IL-1β. Sensory characteristics, such as tactile sensitivity, were found to correlate with IL-6 (p = 0.027) and IL-1β (p = 0.002) in the ASD group. CONCLUSIONS These findings suggest that CMV-infected children with autism are at increased risk of IL-6 and IL-1β dysregulation, contributing to sensory processing issues and eating behaviours. Further research is needed to enhance CMV testing protocols and better understand the virus's role in the development of sensory and behavioural issues in children with autism.
Collapse
Affiliation(s)
- Isti Anindya
- Doctoral Program in Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia;
| | - Rini Sekartini
- Department of Pediatrics, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia;
| | - Ibnu Agus Ariyanto
- Department of Clinical Microbiology, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia;
| | - Tjhin Wiguna
- Department of Psychiatry, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia;
| | - Novika Purnama Sari
- Department Clinical & Developmental Neuropsychology, Faculty of Behavioural and Social Sciences, University of Groningen, 9712 TS Groningen, The Netherlands;
| | | | - Amin Soebandrio
- Department of Clinical Microbiology, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia;
| |
Collapse
|
11
|
Wang B, Qin Y, Chen Y, Zheng X, Chen Y, Zhao J, Zhang F, Duan S. Adipose tissue may not be a major player in the inflammatory pathogenesis of Autism Spectrum Disorder. Brain Behav Immun Health 2025; 43:100929. [PMID: 39810796 PMCID: PMC11732481 DOI: 10.1016/j.bbih.2024.100929] [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: 09/05/2024] [Revised: 12/04/2024] [Accepted: 12/21/2024] [Indexed: 01/16/2025] Open
Abstract
Purpose Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder increasingly recognized for its strong association with chronic inflammation. Adipose tissue functions as an endocrine organ and can secrete inflammatory cytokines to mediate inflammation. However, its involvement in ASD-related inflammation remains unclear. The present study aimed to clarify the role of adipose tissue in inducing inflammatory responses associated with ASD. Methods A total of 36 children with ASD and 18 unrelated healthy controls, aged 2-14.5 years, were enrolled in the study. The up-regulated differentially expressed genes from the GSE18123 dataset were subjected to gene ontology (GO) enrichment analysis to explore ASD-associated pathways. Plasma cytokines and adipokines levels were quantified using Milliplex MAP immunoaffinity technology. The BTBR T + Itprtf/J (BTBR) mice that are known for their core ASD behavioral traits and inflammatory phenotypes were employed as an animal ASD model to verify the key clinical findings. Results GO enrichment analyses revealed immune dysfunction in ASD. Symptom analysis showed that the recruited individuals had typical autistic symptoms. Plasma analysis showed no significant difference in adipokines levels, including adiponectin, leptin, resistin, adipsin, and lipocalin-2, between the ASD and control groups. However, markedly elevated levels of IL-6, IL-8, and tumor necrosis factor (TNF-α) were detected in children with ASD, suggesting that the inflammatory state is independent of adipokines. Similar results were also observed in BTBR autistic mice. Notably, levels of insulin, which are closely related to the exertion of adipokines function, also showed no significant changes. Conclusions Our findings suggest that inflammation in ASD likely originates from non-adipocyte sources, implying that adipose tissue may not play a major role in inflammatory pathogenesis of ASD. Consequently, targeting adipose-related inflammation may not be an effective treatment approach, providing new directions for the development of targeted interventions.
Collapse
Affiliation(s)
- Baojiang Wang
- Institute of Maternal and Child Medicine, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China
- Shenzhen Key Laboratory of Maternal and Child Health and Diseases, Shenzhen, China
| | - Yueyuan Qin
- Institute of Maternal and Child Medicine, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Yong Chen
- Department of Rheumatology and Immunology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xiujie Zheng
- Institute of Maternal and Child Medicine, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Yanjuan Chen
- Department of Rheumatology and Immunology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Juan Zhao
- Institute of Maternal and Child Medicine, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Feng Zhang
- Stomatology Health Care Center, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Shan Duan
- Institute of Maternal and Child Medicine, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China
- Shenzhen Key Laboratory of Maternal and Child Health and Diseases, Shenzhen, China
| |
Collapse
|
12
|
Gidado KI, Adeshakin FO, Rabiu L, Zhang Z, Zhang G, Wan X. Multifaceted roles of DLG3/SAP102 in neurophysiology, neurological disorders and tumorigenesis. Neuroscience 2025; 565:192-201. [PMID: 39638232 DOI: 10.1016/j.neuroscience.2024.11.081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/15/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
DLG3, also known as Synapse-associated protein 102 (SAP102), is essential for the organization and plasticity of excitatory synapses within the central nervous system (CNS). It plays a critical role in clustering and moving key components necessary for learning and memory processes. Mutations in the DLG3 gene, which result in truncated SAP102 proteins, have been associated with a range of neurological disorders, including X-linked intellectual disability (XLID), autism spectrum disorders (ASD), and schizophrenia, all of which can disrupt synaptic structure and cognitive functions. Abnormal SAP102 expression has also been linked to various psychiatric and neurodegenerative conditions, such as bipolar disorder, major depression, and Alzheimer's disease. Recent studies suggest that SAP102 influences cancer development and metastasis by regulating multiple signaling pathways, including the PI3K/AKT axis and the Hippo pathway. Moreover, SAP102 has been demonstrated to regulate tumor-induced bone pain through activating NMDA receptors. These findings highlight SAP102 as a promising therapeutic target for both neurological disorders and cancer. Therefore, further investigation into the regulatory roles of SAP102 in neural development and disease may lead to novel therapeutic approaches for treating synaptic disorders and managing cancer progression.
Collapse
Affiliation(s)
- Khalid Idris Gidado
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Funmilayo O Adeshakin
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Lawan Rabiu
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Ziyang Zhang
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China.
| | - Guizhong Zhang
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China.
| | - Xiaochun Wan
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China.
| |
Collapse
|
13
|
Mallick R, Basak S, Chowdhury P, Bhowmik P, Das RK, Banerjee A, Paul S, Pathak S, Duttaroy AK. Targeting Cytokine-Mediated Inflammation in Brain Disorders: Developing New Treatment Strategies. Pharmaceuticals (Basel) 2025; 18:104. [PMID: 39861166 PMCID: PMC11769149 DOI: 10.3390/ph18010104] [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: 11/22/2024] [Revised: 12/26/2024] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Cytokine-mediated inflammation is increasingly recognized for playing a vital role in the pathophysiology of a wide range of brain disorders, including neurodegenerative, psychiatric, and neurodevelopmental problems. Pro-inflammatory cytokines such as interleukin-1 (IL-1), tumor necrosis factor-alpha (TNF-α), and interleukin-6 (IL-6) cause neuroinflammation, alter brain function, and accelerate disease development. Despite progress in understanding these pathways, effective medicines targeting brain inflammation are still limited. Traditional anti-inflammatory and immunomodulatory drugs are effective in peripheral inflammatory illnesses. Still, they face substantial hurdles when applied to the central nervous system (CNS), such as the blood-brain barrier (BBB) and unwanted systemic effects. This review highlights the developing treatment techniques for modifying cytokine-driven neuroinflammation, focusing on advances that selectively target critical cytokines involved in brain pathology. Novel approaches, including cytokine-specific inhibitors, antibody-based therapeutics, gene- and RNA-based interventions, and sophisticated drug delivery systems like nanoparticles, show promise with respect to lowering neuroinflammation with greater specificity and safety. Furthermore, developments in biomarker discoveries and neuroimaging techniques are improving our ability to monitor inflammatory responses, allowing for more accurate and personalized treatment regimens. Preclinical and clinical trial data demonstrate the therapeutic potential of these tailored techniques. However, significant challenges remain, such as improving delivery across the BBB and reducing off-target effects. As research advances, the creation of personalized, cytokine-centered therapeutics has the potential to alter the therapy landscape for brain illnesses, giving patients hope for better results and a higher quality of life.
Collapse
Affiliation(s)
- Rahul Mallick
- A.I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, 70211 Kuopio, Finland;
| | - Sanjay Basak
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India;
| | - Premanjali Chowdhury
- Institute of Public Health and Clinical Nutrition, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, 70210 Kuopio, Finland;
| | - Prasenjit Bhowmik
- Department of Chemistry, Uppsala Biomedical Centre, Uppsala University, SE-751 23 Uppsala, Sweden;
- Department of Textile Engineering, Green University of Bangladesh, Narayanganj 1461, Bangladesh
| | - Ranjit K. Das
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA;
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Chennai 603103, India; (A.B.); (S.P.)
| | - Sujay Paul
- School of Engineering and Sciences, Tecnologico de Monterrey, Queretaro 76130, Mexico;
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Chennai 603103, India; (A.B.); (S.P.)
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway
| |
Collapse
|
14
|
de Sena Barbosa MG, Vilela de Souza RL, Gonçalves Cherain LG, Santos Ferreira LH, Santos Affi Peixoto ME, Lemos Passos M, Matias Piza L, Almeida Carneiro R, Nunes Rabelo N, Chaurasia B. Biomarkers for autism spectrum disorder: a short review. Ann Med Surg (Lond) 2024; 86:7227-7231. [PMID: 39649921 PMCID: PMC11623892 DOI: 10.1097/ms9.0000000000002689] [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: 06/25/2024] [Accepted: 10/16/2024] [Indexed: 12/11/2024] Open
Abstract
Background Autism spectrum disorder (ASD) is characterized by social disabilities and stereotyped behaviors. There is a relevant social impact on autistic people's lives and, therefore, biomarkers have become relevant for understanding neurobiological mechanisms. Objective This study aims to review current knowledge about the role of biomarkers and their main scientific evidence in autism. Methods The authors performed a non-systematic literature review through the PubMed database, using the keywords "biomarkers", "autism" and "autism spectrum disorder". The search was restricted to articles written in English, in the last 10 years. Results Analyzing the articles found, it is possible to delimit the biomarkers according to the development of ASD, from the prenatal period with exposure to diseases or association of autism with other genetic diseases, through the immune and nutritional factors exposed during pregnancy, and for end those associated with diagnoses phase in which there is the presence of symptoms in which these markers can be used to aid in the diagnosis. Conclusion Although preliminary, biomarkers may hold promise for prenatal and presymptomatic screening. It may also be used as predictors of treatment for autism spectrum disorder.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lívia Matias Piza
- Department of Neurosurgery, Atenas Medical School, Passos, MG, Brazil
| | | | - Nicollas Nunes Rabelo
- Department of Neurosurgery, Atenas Medical School, Passos, MG, Brazil
- Division of Neurosurgery, School of Medicine-University of São Paulo (FMUSP), São Paulo SP, Brazil, Hospital das Clínicas / FMUSP
| | - Bipin Chaurasia
- Department of Neurosurgery, Neurosurgery Clinic, Birgunj, Nepal
| |
Collapse
|
15
|
Wankhede N, Kale M, Shukla M, Nathiya D, R R, Kaur P, Goyanka B, Rahangdale S, Taksande B, Upaganlawar A, Khalid M, Chigurupati S, Umekar M, Kopalli SR, Koppula S. Leveraging AI for the diagnosis and treatment of autism spectrum disorder: Current trends and future prospects. Asian J Psychiatr 2024; 101:104241. [PMID: 39276483 DOI: 10.1016/j.ajp.2024.104241] [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: 06/11/2024] [Revised: 09/05/2024] [Accepted: 09/08/2024] [Indexed: 09/17/2024]
Abstract
The integration of artificial intelligence (AI) into the diagnosis and treatment of autism spectrum disorder (ASD) represents a promising frontier in healthcare. This review explores the current landscape and future prospects of AI technologies in ASD diagnostics and interventions. AI enables early detection and personalized assessment of ASD through the analysis of diverse data sources such as behavioural patterns, neuroimaging, genetics, and electronic health records. Machine learning algorithms exhibit high accuracy in distinguishing ASD from neurotypical development and other developmental disorders, facilitating timely interventions. Furthermore, AI-driven therapeutic interventions, including augmentative communication systems, virtual reality-based training, and robot-assisted therapies, show potential in improving social interactions and communication skills in individuals with ASD. Despite challenges such as data privacy and interpretability, the future of AI in ASD holds promise for refining diagnostic accuracy, deploying telehealth platforms, and tailoring treatment plans. By harnessing AI, clinicians can enhance ASD care delivery, empower patients, and advance our understanding of this complex condition.
Collapse
Affiliation(s)
- Nitu Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Mayur Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Madhu Shukla
- Marwadi University Research Center, Department of Computer Engineering, Faculty of Engineering & Technology, Marwadi University, Rajkot, Gujarat 360003, India
| | - Deepak Nathiya
- Department of Pharmacy Practice, Institute of Pharmacy, NIMS University, Jaipur, India
| | - Roopashree R
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab 140307, India
| | - Barkha Goyanka
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Sandip Rahangdale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Brijesh Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Aman Upaganlawar
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India
| | - Mohammad Khalid
- Department of pharmacognosy, College of pharmacy Prince Sattam Bin Abdulaziz University Alkharj, Saudi Arabia
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraydah 51452, Kingdom of Saudi Arabia
| | - Milind Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea
| |
Collapse
|
16
|
Rezaiezadeh H, Langarizadeh MA, Tavakoli MR, Sabokro M, Banazadeh M, Kohlmeier KA, Shabani M. Therapeutic potential of Bergenin in the management of neurological-based diseases and disorders. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8349-8366. [PMID: 38850305 DOI: 10.1007/s00210-024-03197-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024]
Abstract
Originally sourced from plants, Bergenin has been used as a medicinal compound in traditional medicine for centuries, and anecdotal reports suggest a wide range of therapeutic uses. Naturally-occurring and lab-synthesized Bergenin, as well as some of its related compounds, have been shown in in vivo and in vitro studies to alter activity of several enzymes and proteins critical in cellular functioning, including reelin, GSK-3β, Lingo-1, Ten-4, GP-43, Aβ 1-42, P-tau, SOD1,2, GPx, Glx1, NQO1, HO1, PPAR-ɣ, BDNF, VEGF, and STAT6. Additionally, Bergenin alters levels of several cytokines, such as IL-6, IL-1β, TNF-α, and TGF-β. Behavioral and cellular effects of Bergenin have been shown to involve PI3K/Akt, NF-κB, PKC, Nrf2, and Sirt1/FOXO3a pathways. These pathways, enzymes, and proteins have been shown to be important in normal neurological functioning, and/or dysfunctions in these pathways and proteins have been shown to be important in several neuro-based disorders or diseases, which suggests that Bergenin could be therapeutic in management of neuropsychiatric conditions or neurological disorders. In preclinical studies, Bergenin has been shown to be useful for the management of Alzheimer's disease, Parkinson's disease, anxiety, depression, addiction, epilepsy, insomnia, stroke, and potentially, state control. Our review aims to summarize current evidence supporting the conclusion that Bergenin could play a role in treating various neuro-based disorders and that future studies should be conducted to evaluate the mechanisms by which Bergenin could exert its therapeutic effects.
Collapse
Affiliation(s)
- Hojjat Rezaiezadeh
- Department of Medicinal Chemistry, School of Pharmacy, Shiraz University of Medical Sciences, P.O. Box, Shiraz, 71345-1583, Iran
| | - Mohammad Amin Langarizadeh
- Department of Medicinal Chemistry, Faculty of Pharmacy and Pharmaceutics Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Marziye Ranjbar Tavakoli
- Department of Medicinal Chemistry, Faculty of Pharmacy and Pharmaceutics Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Sabokro
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Banazadeh
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Kristi A Kohlmeier
- Department of Drug Design and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mohammad Shabani
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran.
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
17
|
Szabo A, O'Connell KS, Akkouh IA, Ueland T, Sønderby IE, Hope S, Røe AB, Dønnum MS, Sjaastad I, Steen NE, Ueland T, Sæther LS, Osete JR, Andreassen OA, Nærland T, Djurovic S. Elevated levels of peripheral and central nervous system immune markers reflect innate immune dysregulation in autism spectrum disorder. Psychiatry Res 2024; 342:116245. [PMID: 39481220 DOI: 10.1016/j.psychres.2024.116245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 07/30/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024]
Abstract
BACKGROUND Evidence suggests dysregulated immune functions in the pathophysiology of Autism spectrum disorder (ASD), although specific immune mechanisms are yet to be identified. METHODS We assessed circulating levels of 25 immune/neuroinflammatory markers in a large ASD sample (n = 151) and matched controls (n = 72) using linear models. In addition, we performed global brain transcriptomics analyses of relevant immune-related genes. We also assessed the expression and function of factors and pathway elements of the inflammasome system in peripheral blood mononuclear cells (PBMC) isolated from ASD and controls using in vitro methods. RESULTS We found higher circulating levels of IL-18 and adhesion factors (ICAM-1, MADCAM1) in individuals with ASD relative to controls. Consistent with this, brain levels of ICAM1 mRNA were also higher in ASD compared to controls. Furthermore, we found higher expression/activity of Caspase-1 and the inflammasome sensor NLRP3 in PBMCs in ASD, both at baseline and following inflammatory challenge. This corresponded with higher levels of secreted IL-18, IL-1β, and IL-8, as well as increased expression of adhesion factors following inflammasome activation in ASD PBMC cultures. Inhibition of the NLRP3-inflammasome rescued the observed immune phenotype in ASD in vitro. CONCLUSION Our results suggest a role for inflammasome dysregulation in ASD pathophysiology.
Collapse
Affiliation(s)
- Attila Szabo
- K.G. Jebsen Center for Neurodevelopmental Disorders, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway.
| | - Kevin S O'Connell
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Ibrahim A Akkouh
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, building 25, Kirkeveien 166, Oslo 0450, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Thrombosis Research Center (TREC), Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ida E Sønderby
- K.G. Jebsen Center for Neurodevelopmental Disorders, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, building 25, Kirkeveien 166, Oslo 0450, Norway
| | - Sigrun Hope
- K.G. Jebsen Center for Neurodevelopmental Disorders, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Neurohabilitation, Oslo University Hospital, Oslo, Norway; Department of Rare Disorders and Disabilities, Nevsom, Oslo University Hospital, Oslo, Norway
| | - Anne B Røe
- St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Monica S Dønnum
- Department of Adult Habilitation, Akershus University Hospital, Oslo, Norway
| | - Ingrid Sjaastad
- Department of Child and Adolescent Psychiatry, Vestre Viken Hospital Trust, Norway
| | - Nils Eiel Steen
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Torill Ueland
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Psychology, University of Oslo, Oslo, Norway
| | - Linn Sofie Sæther
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Psychology, University of Oslo, Oslo, Norway
| | - Jordi Requena Osete
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, building 25, Kirkeveien 166, Oslo 0450, Norway
| | - Ole A Andreassen
- K.G. Jebsen Center for Neurodevelopmental Disorders, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Terje Nærland
- K.G. Jebsen Center for Neurodevelopmental Disorders, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Rare Disorders and Disabilities, Nevsom, Oslo University Hospital, Oslo, Norway
| | - Srdjan Djurovic
- K.G. Jebsen Center for Neurodevelopmental Disorders, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, building 25, Kirkeveien 166, Oslo 0450, Norway; Department of Clinical Science, NORMENT, University of Bergen, Bergen, Norway.
| |
Collapse
|
18
|
Fona CMT, Miranda AMM, Jesus MI, Silva VM, Rocha CCS, Costa ACG, Mendes RA. Biomarkers of Pesticide Exposure in a Traditional Brazilian Amazon Community. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:1396. [PMID: 39595663 PMCID: PMC11593667 DOI: 10.3390/ijerph21111396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/14/2024] [Accepted: 10/17/2024] [Indexed: 11/28/2024]
Abstract
In 2008, Brazil became the country with the highest pesticide use in the world, with over one billion liters of pesticides applied to crops in 2009. The impacts of these products on public health are wide-ranging. Vast territories are affected, involving different population groups, such as workers in various fields of activity, the population that consumes contaminated food, and people living around factories, such as traditional communities. This study aimed to assess human exposure to pesticides through epidemiological and laboratory data of residents of the Santo Antônio quilombola community in Concórdia do Pará, Amazon region, Brazil. Epidemiological data were collected using a semi-structured questionnaire, which included factors such as sex, age, length of residence, and level of exposure to pesticides. The modified Ellman method was used to assess the activity of cholinesterases, and flow cytometry was performed for cytokine analysis. Analysis of collected blood samples showed that, in most cases, there was no significant reduction in the activity of acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) compared to other studies in the scientific literature. Meanwhile, there was an increase in the levels of IFN-γ cytokines, especially IL-6, in all groups. The findings of this study highlight the urgent need for a comprehensive monitoring program, considering that some conditions other than pesticide exposure can alter the activities of the biomarkers used in this study.
Collapse
Affiliation(s)
- Cristal M. T. Fona
- Postgraduate Program in Health Surveillance and Epidemiology, Evandro Chagas Institute, Ananindeua 67030-000, Brazil; (C.M.T.F.); (V.M.S.)
| | - Antonio M. M. Miranda
- Environment Section, Evandro Chagas Institute, Ananindeua 67030-000, Brazil; (A.M.M.M.); (M.I.J.); (C.C.S.R.); (A.C.G.C.)
| | - Maria I. Jesus
- Environment Section, Evandro Chagas Institute, Ananindeua 67030-000, Brazil; (A.M.M.M.); (M.I.J.); (C.C.S.R.); (A.C.G.C.)
| | - Viviane M. Silva
- Postgraduate Program in Health Surveillance and Epidemiology, Evandro Chagas Institute, Ananindeua 67030-000, Brazil; (C.M.T.F.); (V.M.S.)
| | - Cássia C. S. Rocha
- Environment Section, Evandro Chagas Institute, Ananindeua 67030-000, Brazil; (A.M.M.M.); (M.I.J.); (C.C.S.R.); (A.C.G.C.)
| | - Amilton C. G. Costa
- Environment Section, Evandro Chagas Institute, Ananindeua 67030-000, Brazil; (A.M.M.M.); (M.I.J.); (C.C.S.R.); (A.C.G.C.)
| | - Rosivaldo A. Mendes
- Environment Section, Evandro Chagas Institute, Ananindeua 67030-000, Brazil; (A.M.M.M.); (M.I.J.); (C.C.S.R.); (A.C.G.C.)
| |
Collapse
|
19
|
King C, Rogers LG, Jansen J, Sivayokan B, Neyhard J, Warnes E, Hall SE, Plakke B. Adolescent treadmill exercise enhances hippocampal brain-derived neurotrophic factor (BDNF) expression and improves cognition in autism-modeled rats. Physiol Behav 2024; 284:114638. [PMID: 39004196 DOI: 10.1016/j.physbeh.2024.114638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/27/2024] [Accepted: 07/09/2024] [Indexed: 07/16/2024]
Abstract
Autism spectrum disorder (ASD) is a prevalent neurodevelopmental disorder characterized by repetitive behaviors and altered communication abilities. Exercise is a low-cost intervention that could improve cognitive function and improve brain plasticity mechanisms. Here, the valproic acid (VPA) model was utilized to induce ASD-like phenotypes in rodents. Animals were exercised on a treadmill and performance was evaluated on a cognitive flexibility task. Biomarkers related to exercise and plasticity regulation were quantified from the prefrontal cortex, hippocampus, and skeletal muscle. Exercised VPA animals had higher levels of hippocampal BDNF compared to sedentary VPA animals and upregulated antioxidant enzyme expression in skeletal muscle. Cognitive improvements were demonstrated in both sexes, but in different domains of cognitive flexibility. This research demonstrates the benefits of exercise and provides evidence that molecular responses to exercise occur in both the central nervous system and in the periphery. These results suggest that improving regulation of BDNF via exercise, even at low intensity, could provide better synaptic regulation and cognitive benefits for individuals with ASD.
Collapse
Affiliation(s)
- Cole King
- Department of Psychological Sciences, Kansas State University, Manhattan, KS, 66506, USA
| | - Liza G Rogers
- Anatomy and Physiology, Kansas State University, Manhattan, KS, 66506, USA
| | - Jeremy Jansen
- Department of Psychological Sciences, Kansas State University, Manhattan, KS, 66506, USA
| | - Bhavana Sivayokan
- Department of Psychological Sciences, Kansas State University, Manhattan, KS, 66506, USA
| | - Jenna Neyhard
- Department of Psychological Sciences, Kansas State University, Manhattan, KS, 66506, USA
| | - Ellie Warnes
- Department of Psychological Sciences, Kansas State University, Manhattan, KS, 66506, USA
| | - Stephanie E Hall
- Anatomy and Physiology, Kansas State University, Manhattan, KS, 66506, USA
| | - Bethany Plakke
- Department of Psychological Sciences, Kansas State University, Manhattan, KS, 66506, USA.
| |
Collapse
|
20
|
King C, Plakke B. Maternal choline supplementation modulates cognition and induces anti-inflammatory signaling in the prefrontal cortices of adolescent rats exposed to maternal immune activation. Brain Behav Immun Health 2024; 40:100836. [PMID: 39206430 PMCID: PMC11350509 DOI: 10.1016/j.bbih.2024.100836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/22/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Maternal infection has long been described as a risk factor for neurodevelopmental disorders, especially autism spectrum disorders (ASD) and schizophrenia. Although many pathogens do not cross the placenta and infect the developing fetus directly, the maternal immune response to them is sufficient to alter fetal neurodevelopment, a phenomenon termed maternal immune activation (MIA). Low maternal choline is also a risk factor for neurodevelopmental disorders, and most pregnant people do not receive enough of it. In addition to its role in neurodevelopment, choline is capable of inducing anti-inflammatory signaling through a nicotinic pathway. Therefore, it was hypothesized that maternal choline supplementation would blunt the neurodevelopmental impact of MIA in offspring through long-term instigation of cholinergic anti-inflammatory signaling. To model MIA in rats, the viral mimetic polyinosinic:polycytidylic acid (poly(I:C)) was used to elicit a maternal antiviral innate immune response in dams both with and without choline supplementation. Offspring were reared to both early and late adolescent stages (postnatal days 28 and 50, respectively), where anxiety-related behaviors and cognition were examined. After behavioral testing, animals were euthanized, and their prefrontal cortices (PFCs) were collected for analysis. MIA offspring demonstrated sex-specific patterns of altered cognition and repetitive behaviors, which were modulated by maternal choline supplementation. Choline supplementation also bolstered anti-inflammatory signaling in the PFCs of MIA animals at both early and late adolescent stages. These findings suggest that maternal choline supplementation may be sufficient to blunt some of the behavioral and neurobiological impacts of inflammatory exposures in utero, indicating that it may be a cheap, safe, and effective intervention for neurodevelopmental disorders.
Collapse
Affiliation(s)
- Cole King
- Department of Psychological Sciences, Kansas State University, 1114 Mid-Campus Drive, Manhattan, KS, 66502, USA
| | - Bethany Plakke
- Department of Psychological Sciences, Kansas State University, 1114 Mid-Campus Drive, Manhattan, KS, 66502, USA
| |
Collapse
|
21
|
Thomas SD, Jayaprakash P, Marwan NZHJ, Aziz EABA, Kuder K, Łażewska D, Kieć-Kononowicz K, Sadek B. Alleviation of Autophagic Deficits and Neuroinflammation by Histamine H3 Receptor Antagonist E159 Ameliorates Autism-Related Behaviors in BTBR Mice. Pharmaceuticals (Basel) 2024; 17:1293. [PMID: 39458934 PMCID: PMC11510413 DOI: 10.3390/ph17101293] [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: 08/09/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Autism spectrum disorder (ASD) is a neurodevelopmental condition marked by social interaction difficulties, repetitive behaviors, and immune dysregulation with elevated pro-inflammatory markers. Autophagic deficiency also contributes to social behavior deficits in ASD. Histamine H3 receptor (H3R) antagonism is a potential treatment strategy for brain disorders with features overlapping ASD, such as schizophrenia and Alzheimer's disease. METHODS This study investigated the effects of sub-chronic systemic treatment with the H3R antagonist E159 on social deficits, repetitive behaviors, neuroinflammation, and autophagic disruption in male BTBR mice. RESULTS E159 (2.5, 5, and 10 mg/kg, i.p.) improved stereotypic repetitive behavior by reducing self-grooming time and enhancing spontaneous alternation in addition to attenuating social deficits. It also decreased pro-inflammatory cytokines in the cerebellum and hippocampus of treated BTBR mice. In BTBR mice, reduced expression of autophagy-related proteins LC3A/B and Beclin 1 was observed, which was elevated following treatment with E159, attenuating the disruption in autophagy. The co-administration with the H3R agonist MHA (10 mg/kg, i.p.) reversed these effects, highlighting the role of histaminergic neurotransmission in observed behavioral improvements. CONCLUSIONS These preliminary findings suggest the therapeutic potential of H3R antagonists in targeting neuroinflammation and autophagic disruption to improve ASD-like behaviors.
Collapse
Affiliation(s)
- Shilu Deepa Thomas
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (S.D.T.); (P.J.); (N.Z.H.J.M.); (E.A.B.A.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Petrilla Jayaprakash
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (S.D.T.); (P.J.); (N.Z.H.J.M.); (E.A.B.A.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Nurfirzana Z. H. J. Marwan
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (S.D.T.); (P.J.); (N.Z.H.J.M.); (E.A.B.A.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Ezzatul A. B. A. Aziz
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (S.D.T.); (P.J.); (N.Z.H.J.M.); (E.A.B.A.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Kamil Kuder
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna Str. 9, 30-688 Kraków, Poland; (K.K.); (D.Ł.); (K.K.-K.)
| | - Dorota Łażewska
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna Str. 9, 30-688 Kraków, Poland; (K.K.); (D.Ł.); (K.K.-K.)
| | - Katarzyna Kieć-Kononowicz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna Str. 9, 30-688 Kraków, Poland; (K.K.); (D.Ł.); (K.K.-K.)
| | - Bassem Sadek
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (S.D.T.); (P.J.); (N.Z.H.J.M.); (E.A.B.A.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
22
|
Anastasescu CM, Gheorman V, Stoicanescu EC, Popescu F, Gheorman V, Udriștoiu I. Immunological Biomarkers in Autism Spectrum Disorder: The Role of TNF-Alpha and Dependent Trends in Serum IL-6 and CXCL8. Life (Basel) 2024; 14:1201. [PMID: 39337983 PMCID: PMC11432970 DOI: 10.3390/life14091201] [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: 07/29/2024] [Revised: 09/12/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) has seen a rise in prevalence, and the immune system's role in brain development is increasingly recognized. This study investigates the relationship between immune dysregulation and ASD by examining serum concentrations of interleukin 6 (IL-6), interleukin 8 (CXCL8), and tumor necrosis factor alpha (TNF-alpha) in children. METHODS Serum samples from 45 children with ASD and 30 controls, aged 2 to 12 years, were analyzed using electrochemiluminescence, chemiluminescent microparticle immunoassay, and chemiluminescent immunoassay. ASD symptoms were assessed using the Autism Spectrum Rating Scale (ASRS) and Social Communication Questionnaire (SCQ). RESULTS No significant correlation was observed between CXCL8 levels and ASD. IL-6 levels showed a trend toward elevation in boys with ASD. TNF-alpha levels were significantly higher in children with ASD under 5 years compared to older children and controls, though no correlation with symptom severity was found. CONCLUSIONS TNF-alpha may be a potential biomarker for early ASD detection, especially in younger children. Further research on larger cohorts is needed to understand the role of immune dysregulation in ASD.
Collapse
Affiliation(s)
| | - Veronica Gheorman
- Department of Medical Semiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Eugen-Cristi Stoicanescu
- Pediatry Department, Emergency Clinical Hospital Râmnicu-Vâlcea, 200300 Râmnicu-Vâlcea, Romania;
| | - Florica Popescu
- Pharmacology Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Victor Gheorman
- Department of Psychiatry, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (V.G.); (I.U.)
| | - Ion Udriștoiu
- Department of Psychiatry, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (V.G.); (I.U.)
| |
Collapse
|
23
|
Kajitani GS, Xavier G, Villena-Rueda BE, Karia BTR, Santoro ML. Extracellular vesicles in neurodegenerative, mental, and other neurological disorders: Perspectives into mechanisms, biomarker potential, and therapeutic implications. CURRENT TOPICS IN MEMBRANES 2024; 94:299-336. [PMID: 39370211 DOI: 10.1016/bs.ctm.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Extracellular vesicles (EVs) are produced, secreted, and targeted by most human cells, including cells that compose nervous system tissues. EVs carry several types of biomolecules, such as lipids, proteins and microRNA, and can function as signaling agents in physiological and pathological processes. In this chapter, we will focus on EVs and their cargo secreted by brain cells, especially neurons and glia, and how these aspects are affected in pathological conditions. The chapter covers neurodegenerative disorders, including Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis, as well as several psychiatric disorders, namely schizophrenia, autism spectrum disorder and major depressive disorder. This chapter also addresses other types of neurological dysfunctions, epilepsy and traumatic brain injury. EVs can cross the blood brain barrier, and thus brain EVs may be detected in more accessible peripheral tissue, such as circulating blood. Alterations in EV composition and contents can therefore impart valuable clues into the molecular etiology of these disorders, and serve biomarkers regarding disease prevalence, progression and treatment. EVs can also be used to carry drugs and biomolecules into brain tissue, considered as a promising drug delivery agent for neurological diseases. Therefore, although this area of research is still in its early development, it offers great potential in further elucidating and in treating neurological disorders.
Collapse
Affiliation(s)
- Gustavo Satoru Kajitani
- Department of Morphology and Genetics, Universidade Federal de São Paulo, Brazil; Laboratory of Integrative Neuroscience, Universidade Federal de São Paulo, Brazil
| | - Gabriela Xavier
- Center for Genomic Medicine, Massachusetts General Hospital, United States; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, United States; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, United States
| | - Beatriz Enguidanos Villena-Rueda
- Department of Morphology and Genetics, Universidade Federal de São Paulo, Brazil; Laboratory of Integrative Neuroscience, Universidade Federal de São Paulo, Brazil
| | - Bruno Takao Real Karia
- Laboratory of Integrative Neuroscience, Universidade Federal de São Paulo, Brazil; Department of Biochemistry, Universidade Federal de São Paulo, Brazil
| | - Marcos Leite Santoro
- Laboratory of Integrative Neuroscience, Universidade Federal de São Paulo, Brazil; Department of Biochemistry, Universidade Federal de São Paulo, Brazil.
| |
Collapse
|
24
|
Wang L, Hu Y, Jiang N, Yetisen AK. Biosensors for psychiatric biomarkers in mental health monitoring. Biosens Bioelectron 2024; 256:116242. [PMID: 38631133 DOI: 10.1016/j.bios.2024.116242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 01/10/2024] [Accepted: 03/22/2024] [Indexed: 04/19/2024]
Abstract
Psychiatric disorders are associated with serve disturbances in cognition, emotional control, and/or behavior regulation, yet few routine clinical tools are available for the real-time evaluation and early-stage diagnosis of mental health. Abnormal levels of relevant biomarkers may imply biological, neurological, and developmental dysfunctions of psychiatric patients. Exploring biosensors that can provide rapid, in-situ, and real-time monitoring of psychiatric biomarkers is therefore vital for prevention, diagnosis, treatment, and prognosis of mental disorders. Recently, psychiatric biosensors with high sensitivity, selectivity, and reproducibility have been widely developed, which are mainly based on electrochemical and optical sensing technologies. This review presented psychiatric disorders with high morbidity, disability, and mortality, followed by describing pathophysiology in a biomarker-implying manner. The latest biosensors developed for the detection of representative psychiatric biomarkers (e.g., cortisol, dopamine, and serotonin) were comprehensively summarized and compared in their sensitivities, sensing technologies, applicable biological platforms, and integrative readouts. These well-developed biosensors are promising for facilitating the clinical utility and commercialization of point-of-care diagnostics. It is anticipated that mental healthcare could be gradually improved in multiple perspectives, ranging from innovations in psychiatric biosensors in terms of biometric elements, transducing principles, and flexible readouts, to the construction of 'Big-Data' networks utilized for sharing intractable psychiatric indicators and cases.
Collapse
Affiliation(s)
- Lin Wang
- Department of Chemical Engineering, Imperial College London, South Kensington, London, SW7 2BU, UK
| | - Yubing Hu
- Department of Chemical Engineering, Imperial College London, South Kensington, London, SW7 2BU, UK.
| | - Nan Jiang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China; Jinfeng Laboratory, Chongqing, 401329, China.
| | - Ali K Yetisen
- Department of Chemical Engineering, Imperial College London, South Kensington, London, SW7 2BU, UK.
| |
Collapse
|
25
|
Kathpalia M, Mishra P, Majid A, Khan MA, Sharma A, Bhurani D, Nidhi. Attenuated adiponectin, omentin, increased interleukin-6 and tumor necrosis factor-alpha levels with altered cognition and depression in non-Hodgkin lymphoma patients: A case-control study. J Neuroimmunol 2024; 392:578372. [PMID: 38788317 DOI: 10.1016/j.jneuroim.2024.578372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/24/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND AND PURPOSE Immune dysfunction is one of the risk factors which plays an important role in the development of non-Hodgkin lymphoma (NHL), and inflammation may be involved in its etiology. Minimal data is available on the effect of cytokine levels on neurobehavioral function in lymphoma before the initiation of chemotherapy. Therefore, we aimed to explore the risk of NHL by assessment of cytokine and adipokine levels and their correlation with neurobehavioral changes. METHODS This case-control study enrolled 62 subjects (age-sex matched: 31 cases and 31 controls). Neurobehavioral assessment was done using Montreal Cognitive Assessment questionnaire (MoCA) and Patient Health Questionnaire (PHQ-9). EORTC Core Quality of Life questionnaire (EORTC QLQ-C30) was used to assess quality of life. Questionnaire assessment and sample collection were done after the patient enrolment and before first cycle of chemotherapy. RESULTS Mean age of NHL patients and healthy controls was 51.9 ± 11.8 and 50 ± 10.9 years, respectively. NHL patients showed significantly higher levels of IL-6 (0.77 ± 0.11) and TNF- α (1.47 ± 1.31) than controls (0.55 ± 0.4 and 0.66 ± 0.89, respectively) with p-value<0.005. Also, NHL patients showed significantly lower levels of adiponectin (0.31 ± 0.24) and omentin (0.46 ± 0.1) than controls (0.42 ± 0.13 and 0.53 ± 0.11, respectively) with p-value<0.005. Lower MoCA and EORTC QLQ C-30 scores and higher PHQ-9 scores were observed in NHL patients in comparison to healthy control. CONCLUSION Our results showed that adiponectin, omentin IL-6 and TNF-α may be used as pre-diagnostic markers of NHL risk. Neurobehavioral changes observed in NHL patients may alter the quality of life.
Collapse
Affiliation(s)
- Meghavi Kathpalia
- Department of Translational and Clinical Research, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, India
| | - Pinki Mishra
- Department of Translational and Clinical Research, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, India
| | - Afsha Majid
- Department of Translational and Clinical Research, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, India
| | - Mohd Ashif Khan
- Department of Translational and Clinical Research, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, India
| | - Anurag Sharma
- Department of Statistics, Ram Lal Anand College, University of Delhi, India
| | - Dinesh Bhurani
- Department of Haemato-oncology and BMT, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Nidhi
- Department of Translational and Clinical Research, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, India.
| |
Collapse
|
26
|
Le Belle JE, Condro M, Cepeda C, Oikonomou KD, Tessema K, Dudley L, Schoenfield J, Kawaguchi R, Geschwind D, Silva AJ, Zhang Z, Shokat K, Harris NG, Kornblum HI. Acute rapamycin treatment reveals novel mechanisms of behavioral, physiological, and functional dysfunction in a maternal inflammation mouse model of autism and sensory over-responsivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602602. [PMID: 39026891 PMCID: PMC11257517 DOI: 10.1101/2024.07.08.602602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Maternal inflammatory response (MIR) during early gestation in mice induces a cascade of physiological and behavioral changes that have been associated with autism spectrum disorder (ASD). In a prior study and the current one, we find that mild MIR results in chronic systemic and neuro-inflammation, mTOR pathway activation, mild brain overgrowth followed by regionally specific volumetric changes, sensory processing dysregulation, and social and repetitive behavior abnormalities. Prior studies of rapamycin treatment in autism models have focused on chronic treatments that might be expected to alter or prevent physical brain changes. Here, we have focused on the acute effects of rapamycin to uncover novel mechanisms of dysfunction and related to mTOR pathway signaling. We find that within 2 hours, rapamycin treatment could rapidly rescue neuronal hyper-excitability, seizure susceptibility, functional network connectivity and brain community structure, and repetitive behaviors and sensory over-responsivity in adult offspring with persistent brain overgrowth. These CNS-mediated effects are also associated with alteration of the expression of several ASD-,ion channel-, and epilepsy-associated genes, in the same time frame. Our findings suggest that mTOR dysregulation in MIR offspring is a key contributor to various levels of brain dysfunction, including neuronal excitability, altered gene expression in multiple cell types, sensory functional network connectivity, and modulation of information flow. However, we demonstrate that the adult MIR brain is also amenable to rapid normalization of these functional changes which results in the rescue of both core and comorbid ASD behaviors in adult animals without requiring long-term physical alterations to the brain. Thus, restoring excitatory/inhibitory imbalance and sensory functional network modularity may be important targets for therapeutically addressing both primary sensory and social behavior phenotypes, and compensatory repetitive behavior phenotypes.
Collapse
|
27
|
Darwish M, El Hajj R, Khayat L, Alaaeddine N. Stem Cell Secretions as a Potential Therapeutic Agent for Autism Spectrum Disorder: A Narrative Review. Stem Cell Rev Rep 2024; 20:1252-1272. [PMID: 38630359 DOI: 10.1007/s12015-024-10724-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2024] [Indexed: 07/04/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental illness characterized by impaired social interaction and restricted repetitive behaviors or interests. The rising prevalence of ASD diagnosis has triggered a surge in research into investigating the underlying neuropathological processes and finding new therapeutic approaches. ASD is characterized by neuroinflammation and dysregulation of neuro-immune cross-talk, which suggests that stem cell treatment might be a potential therapeutic approach. The beneficial and restorative effects of stem cells are mainly due to their paracrine activity, in which stem cells generate and release extracellular vesicles such as exosomes and distinct secreted non-vesicle soluble proteins, including, growth factors, chemokines, cytokines, and immunomodulatory molecules referred to as the Secretome. In this paper, we reviewed the existing research exploring the therapeutic potential of stem cell secretome focusing on their role in addressing ASD pathology. Furthermore, we proposed a comprehensive mechanism of action for stem cell secretions, encompassing the broader secretome as well as the specific contribution of exosomes, in alleviating ASD neuropathology. Across the reviewed studies, exosomes and secreted soluble factors of the transplanted stem cell demonstrate a potential efficacy in ameliorating autistic-like behaviors. The proposed mechanism of action involves the modulation of signaling pathways implicated in neuroinflammation, angiogenesis, cellular apoptosis, and immunomodulation.
Collapse
Affiliation(s)
- Mariam Darwish
- Faculty of Medical Sciences, Neuroscience Research Center, Lebanese University, Beirut, Lebanon
| | | | | | - Nada Alaaeddine
- Dean of Health Sciences, Modern University for Business & Science, Beirut, Lebanon.
| |
Collapse
|
28
|
Chen L, Liu LM, Guo M, Du Y, Chen YW, Xiong XY, Cheng Y. Altered leptin level in autism spectrum disorder and meta-analysis of adipokines. BMC Psychiatry 2024; 24:479. [PMID: 38951775 PMCID: PMC11218410 DOI: 10.1186/s12888-024-05936-4] [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: 03/13/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND Increasing evidence suggests that leptin is involved in the pathology of autism spectrum disorder (ASD). In this study, our objective was to investigate the levels of leptin in the blood of children with ASD and to examine the overall profile of adipokine markers in ASD through meta-analysis. METHODS Leptin concentrations were measured using an enzyme-linked immunosorbent assay (ELISA) kit, while adipokine profiling, including leptin, was performed via meta-analysis. Original reports that included measurements of peripheral adipokines in ASD patients and healthy controls (HCs) were collected from databases such as Web of Science, PubMed, and Cochrane Library. These studies were collected from September 2022 to September 2023 and followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Standardized mean differences were calculated using a random effects model for the meta-analysis. Additionally, we performed meta-regression and explored heterogeneity among studies. RESULTS Our findings revealed a significant increase in leptin levels in children with ASD compared to HCs (p = 0.0319). This result was consistent with the findings obtained from the meta-analysis (p < 0.001). Furthermore, progranulin concentrations were significantly reduced in children with ASD. However, for the other five adipokines analyzed, there were no significant differences observed between the children with ASD and HCs children. Heterogeneity was found among the studies, and the meta-regression analysis indicated that publication year and latitude might influence the results of the meta-analysis. CONCLUSIONS These findings provide compelling evidence that leptin levels are increased in children with ASD compared to healthy controls, suggesting a potential mechanism involving adipokines, particularly leptin, in the pathogenesis of ASD. These results contribute to a better understanding of the pathology of ASD and provide new insights for future investigations.
Collapse
Affiliation(s)
- Lei Chen
- Key Laboratory of Ethnomedicine of Ministry of Education, Center On Translational Neuroscience, School of Pharmacy, Minzu University of China, 27 South Zhongguancun Avenue, Beijing, 100081, China
| | - Li-Ming Liu
- Institute of National Security, Minzu University of China, Beijing, China
| | - Mei Guo
- Key Laboratory of Ethnomedicine of Ministry of Education, Center On Translational Neuroscience, School of Pharmacy, Minzu University of China, 27 South Zhongguancun Avenue, Beijing, 100081, China
| | - Yang Du
- Key Laboratory of Ethnomedicine of Ministry of Education, Center On Translational Neuroscience, School of Pharmacy, Minzu University of China, 27 South Zhongguancun Avenue, Beijing, 100081, China
| | - Yue-Wen Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Key Laboratory of Translational Research for Brain Diseases, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen,, 518055, Guangdong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, 518057, Guangdong, China
| | - Xi-Yue Xiong
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, China.
| | - Yong Cheng
- Key Laboratory of Ethnomedicine of Ministry of Education, Center On Translational Neuroscience, School of Pharmacy, Minzu University of China, 27 South Zhongguancun Avenue, Beijing, 100081, China.
- Institute of National Security, Minzu University of China, Beijing, China.
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, China.
| |
Collapse
|
29
|
Nautiyal H, Jaiswar A, Jha PK, Dwivedi S. Exploring key genes and pathways associated with sex differences in autism spectrum disorder: integrated bioinformatic analysis. Mamm Genome 2024; 35:280-295. [PMID: 38594551 DOI: 10.1007/s00335-024-10036-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 02/20/2024] [Indexed: 04/11/2024]
Abstract
Autism spectrum disorder (ASD) is a heterogenous neurodevelopmental disorder marked by functional abnormalities in brain that causes social and linguistic difficulties. The incidence of ASD is more prevalent in males compared to females, but the underlying mechanism, as well as molecular indications for identifying sex-specific differences in ASD symptoms remain unknown. Thus, impacting the development of personalized strategy towards pharmacotherapy of ASD. The current study employs an integrated bioinformatic approach to investigate the genes and pathways uniquely associated with sex specific differences in autistic individuals. Based on microarray dataset (GSE6575) extracted from the gene expression omnibus, the dysregulated genes between the autistic and the neurotypical individuals for both sexes were identified. Gene set enrichment analysis was performed to ascertain biological activities linked to the dysregulated genes. Protein-protein interaction network analysis was carried out to identify hub genes. The identified hub genes were examined to determine their functions and involvement in the associated pathways using Enrichr. Additionally, hub genes were validated from autism-associated databases and the potential small molecules targeting the hub genes were identified. The present study utilized whole blood transcriptomic gene expression analysis data and identified 2211 and 958 differentially expressed unique genes in males and females respectively. The functional enrichment analysis revealed that male hub genes were functionally associated with RNA polymerase II mediated transcriptional regulation whereas female hub genes were involved in intracellular signal transduction and cell migration. The top male hub genes exhibited functional enrichment in tyrosine kinase signalling pathway. The pathway enrichment analysis of male hub genes indicates the enrichment of papillomavirus infection. Female hub genes were enriched in androgen receptor signalling pathway and functionally enriched in focal adhesion specific excision repair. Identified drug like candidates targeting these genes may serve as a potential sex specific therapeutics. Wortmannin for males, 5-Fluorouracil for females had the highest scores. Targeted and sex-specific pharmacotherapies may be created for the management of ASD. The current investigation identifies sex-specific molecular signatures derived from whole blood which may serve as a potential peripheral sex-specific biomarkers for ASD. The study also uncovers the possible pharmacological interventions against the selected genes/pathway, providing support in development of therapeutic strategies to mitigate ASD. However, experimental proofs on biological systems are warranted.
Collapse
Affiliation(s)
- Himani Nautiyal
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, 248001, India
| | - Akanksha Jaiswar
- Laboratory of Human Disease Multiomics, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
| | - Prabhash Kumar Jha
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shubham Dwivedi
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, 248001, India.
| |
Collapse
|
30
|
Osman HC, Moreno R, Rose D, Rowland ME, Ciernia AV, Ashwood P. Impact of maternal immune activation and sex on placental and fetal brain cytokine and gene expression profiles in a preclinical model of neurodevelopmental disorders. J Neuroinflammation 2024; 21:118. [PMID: 38715090 PMCID: PMC11077729 DOI: 10.1186/s12974-024-03106-7] [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: 12/21/2023] [Accepted: 04/21/2024] [Indexed: 05/12/2024] Open
Abstract
Maternal inflammation during gestation is associated with a later diagnosis of neurodevelopmental disorders including autism spectrum disorder (ASD). However, the specific impact of maternal immune activation (MIA) on placental and fetal brain development remains insufficiently understood. This study aimed to investigate the effects of MIA by analyzing placental and brain tissues obtained from the offspring of pregnant C57BL/6 dams exposed to polyinosinic: polycytidylic acid (poly I: C) on embryonic day 12.5. Cytokine and mRNA content in the placenta and brain tissues were assessed using multiplex cytokine assays and bulk-RNA sequencing on embryonic day 17.5. In the placenta, male MIA offspring exhibited higher levels of GM-CSF, IL-6, TNFα, and LT-α, but there were no differences in female MIA offspring. Furthermore, differentially expressed genes (DEG) in the placental tissues of MIA offspring were found to be enriched in processes related to synaptic vesicles and neuronal development. Placental mRNA from male and female MIA offspring were both enriched in synaptic and neuronal development terms, whereas females were also enriched for terms related to excitatory and inhibitory signaling. In the fetal brain of MIA offspring, increased levels of IL-28B and IL-25 were observed with male MIA offspring and increased levels of LT-α were observed in the female offspring. Notably, we identified few stable MIA fetal brain DEG, with no male specific difference whereas females had DEG related to immune cytokine signaling. Overall, these findings support the hypothesis that MIA contributes to the sex- specific abnormalities observed in ASD, possibly through altered neuron developed from exposure to inflammatory cytokines. Future research should aim to investigate how interactions between the placenta and fetal brain contribute to altered neuronal development in the context of MIA.
Collapse
Affiliation(s)
- Hadley C Osman
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, USA
- The M.I.N.D. Institute, University of California at Sacramento, Sacramento, CA, USA
| | - Rachel Moreno
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, USA
- The M.I.N.D. Institute, University of California at Sacramento, Sacramento, CA, USA
| | - Destanie Rose
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, USA
- The M.I.N.D. Institute, University of California at Sacramento, Sacramento, CA, USA
| | - Megan E Rowland
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Annie Vogel Ciernia
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, USA.
- The M.I.N.D. Institute, University of California at Sacramento, Sacramento, CA, USA.
| |
Collapse
|
31
|
Allan NP, Yamamoto BY, Kunihiro BP, Nunokawa CKL, Rubas NC, Wells RK, Umeda L, Phankitnirundorn K, Torres A, Peres R, Takahashi E, Maunakea AK. Ketogenic Diet Induced Shifts in the Gut Microbiome Associate with Changes to Inflammatory Cytokines and Brain-Related miRNAs in Children with Autism Spectrum Disorder. Nutrients 2024; 16:1401. [PMID: 38794639 PMCID: PMC11124410 DOI: 10.3390/nu16101401] [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: 04/04/2024] [Revised: 05/01/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
In this interventional pilot study, we investigated the effects of a modified ketogenic diet (KD) on children with autism spectrum disorder (ASD). We previously observed improved behavioral symptoms in this cohort following the KD; this trial was registered with Clinicaltrials.gov (NCT02477904). This report details the alterations observed in the microbiota, inflammation markers, and microRNAs of seven children following a KD for a duration of 4 months. Our analysis included blood and stool samples, collected before and after the KD. After 4 months follow up, we found that the KD led to decreased plasma levels of proinflammatory cytokines (IL-12p70 and IL-1b) and brain-derived neurotrophic factor (BDNF). Additionally, we observed changes in the gut microbiome, increased expression of butyrate kinase in the gut, and altered levels of BDNF-associated miRNAs in the plasma. These cohort findings suggest that the KD may positively influence ASD sociability, as previously observed, by reducing inflammation, reversing gut microbial dysbiosis, and impacting the BDNF pathway related to brain activity.
Collapse
Affiliation(s)
- Nina P. Allan
- Department of Biochemistry, Anatomy, and Physiology, University of Hawai’i at Mānoa, Honolulu, HI 96822, USA; (N.P.A.); (B.Y.Y.); (B.P.K.); (C.K.L.N.); (N.C.R.); (R.K.W.); (L.U.); (K.P.); (A.T.); (R.P.)
| | - Brennan Y. Yamamoto
- Department of Biochemistry, Anatomy, and Physiology, University of Hawai’i at Mānoa, Honolulu, HI 96822, USA; (N.P.A.); (B.Y.Y.); (B.P.K.); (C.K.L.N.); (N.C.R.); (R.K.W.); (L.U.); (K.P.); (A.T.); (R.P.)
| | - Braden P. Kunihiro
- Department of Biochemistry, Anatomy, and Physiology, University of Hawai’i at Mānoa, Honolulu, HI 96822, USA; (N.P.A.); (B.Y.Y.); (B.P.K.); (C.K.L.N.); (N.C.R.); (R.K.W.); (L.U.); (K.P.); (A.T.); (R.P.)
| | - Chandler K. L. Nunokawa
- Department of Biochemistry, Anatomy, and Physiology, University of Hawai’i at Mānoa, Honolulu, HI 96822, USA; (N.P.A.); (B.Y.Y.); (B.P.K.); (C.K.L.N.); (N.C.R.); (R.K.W.); (L.U.); (K.P.); (A.T.); (R.P.)
| | - Noelle C. Rubas
- Department of Biochemistry, Anatomy, and Physiology, University of Hawai’i at Mānoa, Honolulu, HI 96822, USA; (N.P.A.); (B.Y.Y.); (B.P.K.); (C.K.L.N.); (N.C.R.); (R.K.W.); (L.U.); (K.P.); (A.T.); (R.P.)
- Molecular Biosciences and Bioengineering, College of Tropical Agriculture and Human Resources, University of Hawai’i at Manoa, Honolulu, HI 96822, USA
| | - Riley K. Wells
- Department of Biochemistry, Anatomy, and Physiology, University of Hawai’i at Mānoa, Honolulu, HI 96822, USA; (N.P.A.); (B.Y.Y.); (B.P.K.); (C.K.L.N.); (N.C.R.); (R.K.W.); (L.U.); (K.P.); (A.T.); (R.P.)
- Molecular Biosciences and Bioengineering, College of Tropical Agriculture and Human Resources, University of Hawai’i at Manoa, Honolulu, HI 96822, USA
| | - Lesley Umeda
- Department of Biochemistry, Anatomy, and Physiology, University of Hawai’i at Mānoa, Honolulu, HI 96822, USA; (N.P.A.); (B.Y.Y.); (B.P.K.); (C.K.L.N.); (N.C.R.); (R.K.W.); (L.U.); (K.P.); (A.T.); (R.P.)
- Molecular Biosciences and Bioengineering, College of Tropical Agriculture and Human Resources, University of Hawai’i at Manoa, Honolulu, HI 96822, USA
| | - Krit Phankitnirundorn
- Department of Biochemistry, Anatomy, and Physiology, University of Hawai’i at Mānoa, Honolulu, HI 96822, USA; (N.P.A.); (B.Y.Y.); (B.P.K.); (C.K.L.N.); (N.C.R.); (R.K.W.); (L.U.); (K.P.); (A.T.); (R.P.)
| | - Amada Torres
- Department of Biochemistry, Anatomy, and Physiology, University of Hawai’i at Mānoa, Honolulu, HI 96822, USA; (N.P.A.); (B.Y.Y.); (B.P.K.); (C.K.L.N.); (N.C.R.); (R.K.W.); (L.U.); (K.P.); (A.T.); (R.P.)
| | - Rafael Peres
- Department of Biochemistry, Anatomy, and Physiology, University of Hawai’i at Mānoa, Honolulu, HI 96822, USA; (N.P.A.); (B.Y.Y.); (B.P.K.); (C.K.L.N.); (N.C.R.); (R.K.W.); (L.U.); (K.P.); (A.T.); (R.P.)
| | - Emi Takahashi
- Department of Radiology, Harvard Medical School, Boston, MA 02115, USA;
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Alika K. Maunakea
- Department of Biochemistry, Anatomy, and Physiology, University of Hawai’i at Mānoa, Honolulu, HI 96822, USA; (N.P.A.); (B.Y.Y.); (B.P.K.); (C.K.L.N.); (N.C.R.); (R.K.W.); (L.U.); (K.P.); (A.T.); (R.P.)
| |
Collapse
|
32
|
Borreca A, Mantovani C, Desiato G, Corradini I, Filipello F, Elia CA, D'Autilia F, Santamaria G, Garlanda C, Morini R, Pozzi D, Matteoli M. Loss of interleukin 1 signaling causes impairment of microglia- mediated synapse elimination and autistic-like behaviour in mice. Brain Behav Immun 2024; 117:493-509. [PMID: 38307446 DOI: 10.1016/j.bbi.2024.01.221] [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: 10/05/2023] [Revised: 01/11/2024] [Accepted: 01/25/2024] [Indexed: 02/04/2024] Open
Abstract
In the last years, the hypothesis that elevated levels of proinflammatory cytokines contribute to the pathogenesis of neurodevelopmental diseases has gained popularity. IL-1 is one of the main cytokines found to be elevated in Autism spectrum disorder (ASD), a complex neurodevelopmental condition characterized by defects in social communication and cognitive impairments. In this study, we demonstrate that mice lacking IL-1 signaling display autistic-like defects associated with an excessive number of synapses. We also show that microglia lacking IL-1 signaling at early neurodevelopmental stages are unable to properly perform the process of synapse engulfment and display excessive activation of mammalian target of rapamycin (mTOR) signaling. Notably, even the acute inhibition of IL-1R1 by IL-1Ra is sufficient to enhance mTOR signaling and reduce synaptosome phagocytosis in WT microglia. Finally, we demonstrate that rapamycin treatment rescues the defects in IL-1R deficient mice. These data unveil an exclusive role of microglial IL-1 in synapse refinement via mTOR signaling and indicate a novel mechanism possibly involved in neurodevelopmental disorders associated with defects in the IL-1 pathway.
Collapse
Affiliation(s)
- Antonella Borreca
- Institute of Neuroscience (IN-CNR), Consiglio Nazionale delle Ricerche, Milan, Italy; IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Cristina Mantovani
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy
| | - Genni Desiato
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Irene Corradini
- Institute of Neuroscience (IN-CNR), Consiglio Nazionale delle Ricerche, Milan, Italy; IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Fabia Filipello
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Chiara Adriana Elia
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Francesca D'Autilia
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Giulia Santamaria
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Cecilia Garlanda
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy
| | - Raffaella Morini
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Davide Pozzi
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy.
| | - Michela Matteoli
- Institute of Neuroscience (IN-CNR), Consiglio Nazionale delle Ricerche, Milan, Italy; IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy.
| |
Collapse
|
33
|
Kovacheva E, Gevezova M, Maes M, Sarafian V. Mast Cells in Autism Spectrum Disorder-The Enigma to Be Solved? Int J Mol Sci 2024; 25:2651. [PMID: 38473898 DOI: 10.3390/ijms25052651] [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: 01/15/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Autism Spectrum Disorder (ASD) is a disturbance of neurodevelopment with a complicated pathogenesis and unidentified etiology. Many children with ASD have a history of "allergic symptoms", often in the absence of mast cell (MC)-positive tests. Activation of MCs by various stimuli may release molecules related to inflammation and neurotoxicity, contributing to the development of ASD. The aim of the present paper is to enrich the current knowledge on the relationship between MCs and ASD by discussing key molecules and immune pathways associated with MCs in the pathogenesis of autism. Cytokines, essential marker molecules for MC degranulation and therapeutic targets, are also highlighted. Understanding the relationship between ASD and the activation of MCs, as well as the involved molecules and interactions, are the main points contributing to solving the enigma. Key molecules, associated with MCs, may provide new insights to the discovery of drug targets for modeling inflammation in ASD.
Collapse
Affiliation(s)
- Eleonora Kovacheva
- Department of Medical Biology, Medical University-Plovdiv, 4000 Plovdiv, Bulgaria
- Research Institute, Medical University-Plovdiv, 4000 Plovdiv, Bulgaria
| | - Maria Gevezova
- Department of Medical Biology, Medical University-Plovdiv, 4000 Plovdiv, Bulgaria
- Research Institute, Medical University-Plovdiv, 4000 Plovdiv, Bulgaria
| | - Michael Maes
- Research Institute, Medical University-Plovdiv, 4000 Plovdiv, Bulgaria
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu 610072, China
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Cognitive Fitness and Technology Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Psychiatry, Medical University-Plovdiv, 4000 Plovdiv, Bulgaria
- Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Victoria Sarafian
- Department of Medical Biology, Medical University-Plovdiv, 4000 Plovdiv, Bulgaria
- Research Institute, Medical University-Plovdiv, 4000 Plovdiv, Bulgaria
| |
Collapse
|
34
|
Suprunowicz M, Tomaszek N, Urbaniak A, Zackiewicz K, Modzelewski S, Waszkiewicz N. Between Dysbiosis, Maternal Immune Activation and Autism: Is There a Common Pathway? Nutrients 2024; 16:549. [PMID: 38398873 PMCID: PMC10891846 DOI: 10.3390/nu16040549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/05/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
Autism spectrum disorder (ASD) is a neuropsychiatric condition characterized by impaired social interactions and repetitive stereotyped behaviors. Growing evidence highlights an important role of the gut-brain-microbiome axis in the pathogenesis of ASD. Research indicates an abnormal composition of the gut microbiome and the potential involvement of bacterial molecules in neuroinflammation and brain development disruptions. Concurrently, attention is directed towards the role of short-chain fatty acids (SCFAs) and impaired intestinal tightness. This comprehensive review emphasizes the potential impact of maternal gut microbiota changes on the development of autism in children, especially considering maternal immune activation (MIA). The following paper evaluates the impact of the birth route on the colonization of the child with bacteria in the first weeks of life. Furthermore, it explores the role of pro-inflammatory cytokines, such as IL-6 and IL-17a and mother's obesity as potentially environmental factors of ASD. The purpose of this review is to advance our understanding of ASD pathogenesis, while also searching for the positive implications of the latest therapies, such as probiotics, prebiotics or fecal microbiota transplantation, targeting the gut microbiota and reducing inflammation. This review aims to provide valuable insights that could instruct future studies and treatments for individuals affected by ASD.
Collapse
Affiliation(s)
| | | | | | | | - Stefan Modzelewski
- Department of Psychiatry, Medical University of Bialystok, pl. Wołodyjowskiego 2, 15-272 Białystok, Poland; (M.S.); (N.T.); (A.U.); (K.Z.); (N.W.)
| | | |
Collapse
|
35
|
Santos TCD, Obando JMC, Leite PEC, Pereira MR, Leitão MDF, Abujadi C, Pimenta LDFL, Martins RCC, Cavalcanti DN. Approaches of marine compounds and relevant immune mediators in Autism Spectrum Disorder: Opportunities and challenges. Eur J Med Chem 2024; 266:116153. [PMID: 38277916 DOI: 10.1016/j.ejmech.2024.116153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/12/2024] [Accepted: 01/13/2024] [Indexed: 01/28/2024]
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder that affects social skills, language, communication, and behavioral skills, significantly impacting the individual's quality of life. Recently, numerous works have centered on the connections between the immune and central nervous systems and the influence of neuroinflammation on autism symptomatology. Marine natural products are considered as important alternative sources of different types of compounds, including polysaccharides, polyphenols, sterols, carotenoids, terpenoids and, alkaloids. These compounds present anti-inflammatory, neuroprotective and immunomodulatory activities, exhibiting a potential for the treatment of many diseases. Although many studies address the marine compounds in the modulation of inflammatory mediators, there is a gap regarding their use in the regulation of the immune system in ASD. Thus, this review aims to provide a better understanding regarding cytokines, chemokines, growth factors and immune responses in ASD, as well as the potential of bioactive marine compounds in the immune regulation in ASD. We expect that this review would contribute to the development of therapeutic alternatives for controlling immune mediators and inflammation in ASD.
Collapse
Affiliation(s)
- Thalisia Cunha Dos Santos
- Programa de Pós-graduação em Química de Produtos Naturais, Instituto de Pesquisas de Produtos Naturais Walter Mors, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Laboratório de Produtos Naturais de Algas Marinha (ALGAMAR), Departamento de Biologia Marinha, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Núcleo de Estudos e Pesquisas em Autismo (NEPA), Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil.
| | - Johana Marcela Concha Obando
- Laboratório de Produtos Naturais de Algas Marinha (ALGAMAR), Departamento de Biologia Marinha, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Núcleo de Estudos e Pesquisas em Autismo (NEPA), Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Paulo Emílio Corrêa Leite
- Núcleo de Estudos e Pesquisas em Autismo (NEPA), Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Programa de Pós-graduação em Ciências e Biotecnologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Instituto LisMAPS, Niterói, RJ, Brazil
| | - Mariana Rodrigues Pereira
- Programa de Pós-graduação em Ciências e Biotecnologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Instituto LisMAPS, Niterói, RJ, Brazil; Programa de Pós-graduação em Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Mônica de Freitas Leitão
- Núcleo de Estudos e Pesquisas em Autismo (NEPA), Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Faculdade de Medicina, Pontifícia Universidade Católica de Campinas (PUC-Camp), Campinas, SP, Brazil
| | - Caio Abujadi
- Núcleo de Estudos e Pesquisas em Autismo (NEPA), Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Programa de Pós-graduação em Ciência, Tecnologia e Inclusão (PGCTIn), Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | | | - Roberto Carlos Campos Martins
- Programa de Pós-graduação em Química de Produtos Naturais, Instituto de Pesquisas de Produtos Naturais Walter Mors, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Diana Negrão Cavalcanti
- Laboratório de Produtos Naturais de Algas Marinha (ALGAMAR), Departamento de Biologia Marinha, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Núcleo de Estudos e Pesquisas em Autismo (NEPA), Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Programa de Pós-graduação em Ciência, Tecnologia e Inclusão (PGCTIn), Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil.
| |
Collapse
|
36
|
Akintunde ME, Lin YP, Krakowiak P, Pessah IN, Hertz-Picciotto I, Puschner B, Ashwood P, Van de Water J. Ex vivo exposure to polybrominated diphenyl ether (PBDE) selectively affects the immune response in autistic children. Brain Behav Immun Health 2023; 34:100697. [PMID: 38020477 PMCID: PMC10654005 DOI: 10.1016/j.bbih.2023.100697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/21/2023] [Accepted: 10/22/2023] [Indexed: 12/01/2023] Open
Abstract
Children on the autism spectrum have been shown to have immune dysregulation that often correlates with behavioral deficits. The role of the post-natal environment in this dysregulation is an area of active investigation. We examined the association between plasma levels of polybrominated diphenyl ether (PBDE) and immune cell function in age-matched autistic children and non-autistic controls. Plasma from children on the autism spectrum (n = 38) and typically developing controls (TD; n = 60) were analyzed for 14 major PBDE congeners. Cytokine/chemokine production was measured in peripheral blood mononuclear cell (PBMC) supernatants with and without ex vivo BDE-49 exposure. Total plasma concentration (∑PBDE14) and individual congener levels were also correlated with T cell function. ∑PBDE14 did not differ between diagnostic groups but correlated with reduced immune function in children on the autism spectrum. In autistic children, IL-2 and IFN-γ production was reduced in association with several individual BDE congeners, especially BDE-49 (p = 0.001). Furthermore, when PBMCs were exposed ex vivo to BDE-49, cells from autistic children produced elevated levels of IL-6, TNF-α, IL-1β, MIP-1α and MCP-1 (p < 0.05). Therefore, despite similar plasma levels of PBDE, these data suggest that PBMC function was differentially impacted in the context of several PBDE congeners in autistic children relative to TD children where increased body burden of PBDE significantly correlated with a suppressed immune response in autistic children but not TD controls. Further, acute ex vivo exposure of PBMCs to BDE-49 stimulates an elevated cytokine response in AU cases versus a depressed response in TD controls. These data suggest that exposure to the toxicant BDE-49 differentially impacts immune cell function in autistic children relative to TD children providing evidence for an underlying association between susceptibility to PBDE exposure and immune anomalies in children on the autism spectrum.
Collapse
Affiliation(s)
- Marjannie Eloi Akintunde
- School of Medicine, Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, United States
- NIEHS Center for Children's Environmental Health, University of California, Davis, United States
| | - Yan-ping Lin
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, United States
- The MIND Institute, University of California, Davis, United States
- NIEHS Center for Children's Environmental Health, University of California, Davis, United States
| | - Paula Krakowiak
- The MIND Institute, University of California, Davis, United States
- School of Public Health Sciences, University of California, Davis, United States
| | - Isaac N. Pessah
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, United States
- The MIND Institute, University of California, Davis, United States
- NIEHS Center for Children's Environmental Health, University of California, Davis, United States
| | - Irva Hertz-Picciotto
- The MIND Institute, University of California, Davis, United States
- School of Public Health Sciences, University of California, Davis, United States
| | - Birgit Puschner
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, United States
- NIEHS Center for Children's Environmental Health, University of California, Davis, United States
| | - Paul Ashwood
- The MIND Institute, University of California, Davis, United States
- NIEHS Center for Children's Environmental Health, University of California, Davis, United States
- School of Medicine, Department of Microbiology and Immunology, University of California, Davis, United States
| | - Judy Van de Water
- School of Medicine, Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, United States
- The MIND Institute, University of California, Davis, United States
- NIEHS Center for Children's Environmental Health, University of California, Davis, United States
| |
Collapse
|
37
|
Halepoto DM, AL-Ayadhi LY, Alhowikan AM, Elamin NE. Role of autoimmunity in Neuronal damage in children with Autism spectrum disorder. Pak J Med Sci 2023; 39:1858-1864. [PMID: 37936741 PMCID: PMC10626070 DOI: 10.12669/pjms.39.6.6804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 04/10/2023] [Accepted: 04/24/2023] [Indexed: 11/09/2023] Open
Abstract
"Autism spectrum disorder (ASD) is complex neurodevelopmental disorder characterized by impairments in three core behavioral: social deficits, impaired communication, and repetitive behaviors." There is developing indication and emerging data that irregular autoimmune responses to the central nervous system may play a pathogenic role in patients with autism spectrum disorder." The aim of this review was to discuss the updated research carried out at Autism research and treatment center, King Saud University, Riyadh, Kingdom of Saudi Arabia particularly on the role of autoimmunity in Autism spectrum disorder. This review also present state of information available about the role of autoimmunity biomarkers involved in the neuronal damage of central nervous system in autistic children. The systematic literature search was carried out using Google Scholar, Science direct and PubMed databases on the role of autoimmunity in autism and reviewed all relevant articles published in peer reviewed journals by Autism research and treatment center, King Saud University, Riyadh, Kingdom of Saudi Arabia till April, 2022. We searched relevant articles using key words Autism spectrum disorder, Autoimmunity, Neuroinflamation and Central nervous system. This review revealed that plasma levels of autoimmunity related factors/ markers were altered in patients with autism. Significant change in blood markers in subjects with ASD may resulted in several years of decreased neutrotrophic support along with increasing impairment in relationship with down-regulated inflammation that may play a role in the ASD. Overall, the role of autoimmunity in ASD subjects with excess of anti-brain antibodies suggest that in some patients, autoantibodies that target the CNS may be pathological factor in neuronal growth in autistic children. Large cohort studies with well-defined and specially pheno typed autistic groups and matched healthy controls are required to examine the role of autoantibodies in the pathology of subjects with ASD.
Collapse
Affiliation(s)
- Dost Muhammad Halepoto
- Dr. Dost Muhammad Halepoto, PhD. Autism Research and Treatment center, Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Laila Yousif AL-Ayadhi
- Dr. Laila Yousif AL-Ayadhi, PhD. Autism Research and Treatment center, Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Abdulrahman Mohammed Alhowikan
- Dr. Abdulrahman Mohammed Alhowikan, PhD. Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Nadra Elyass Elamin
- Dr. Nadra Elyass Elamin, PhD. Autism Research and Treatment Center, Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
38
|
Al-Mazidi SH. The Physiology of Cognition in Autism Spectrum Disorder: Current and Future Challenges. Cureus 2023; 15:e46581. [PMID: 37808604 PMCID: PMC10557542 DOI: 10.7759/cureus.46581] [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] [Accepted: 10/06/2023] [Indexed: 10/10/2023] Open
Abstract
Cognitive impairment is among the most challenging characteristics of autism spectrum disorder (ASD). Although ASD is one of the common neurodevelopmental disorders, we are still behind in diagnosing and treating cognitive impairment in ASD. Cognitive impairment in ASD varies, meaning it could be at the sensory perception level to cognitive processing, learning, and memory. There are no diagnostic criteria for cognitive impairment that are specific to ASD. The leading causes of cognitive impairment in ASD could be neurological, immune, and gastrointestinal dysfunction. Immune dysfunction might lead to neuroinflammation, affecting neural connectivity, glutamate/gamma-aminobutyric acid (GABA) balance, and plasticity. The gut-brain axes are essential in the developing brain. Special retinal changes have recently been detected in ASD, which need clinical investigation to find their possible role in early diagnosis. Early intervention is crucial for ASD cognitive dysfunction. Due to the heterogeneity of the disease, the clinical manifestation of ASD makes it difficult for clinicians to develop gold-standard diagnostic and therapeutic criteria. We suggest a triad for diagnosis, which includes clinical tests for immune and gastrointestinal dysfunction biomarkers, clinical examination for the retina, and an objective neurocognitive evaluation for ASD, and to develop a treatment strategy involving these three aspects. Developing clear treatment criteria for cognitive impairment for ASD would improve the quality of life of ASD people and their caregivers and would delay or prevent dementia-related disorders in ASD people.
Collapse
|
39
|
Zhang L, Xu Y, Sun S, Liang C, Li W, Li H, Zhang X, Pang D, Li M, Li H, Lang Y, Liu J, Jiang S, Shi X, Li B, Yang Y, Wang Y, Li Z, Song C, Duan G, Leavenworth JW, Wang X, Zhu C. Integrative analysis of γδT cells and dietary factors reveals predictive values for autism spectrum disorder in children. Brain Behav Immun 2023; 111:76-89. [PMID: 37011865 DOI: 10.1016/j.bbi.2023.03.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) includes a range of multifactorial neurodevelopmental disabilities characterized by a variable set of neuropsychiatric symptoms. Immunological abnormalities have been considered to play important roles in the pathogenesis of ASD, but it is still unknown which abnormalities are more prominent. METHODS A total of 105 children with ASD and 105 age and gender-matched typically developing (TD) children were recruited. An eating and mealtime behavior questionnaire, dietary habits, and the Bristol Stool Scale were investigated. The immune cell profiles in peripheral blood were analyzed by flow cytometry, and cytokines (IFN-γ, IL-8, IL-10, IL-17A, and TNF-α) in plasma were examined by Luminex assay. The obtained results were further validated using an external validation cohort including 82 children with ASD and 51 TD children. RESULTS Compared to TD children, children with ASD had significant eating and mealtime behavioral changes and gastrointestinal symptoms characterized by increased food fussiness and emotional eating, decreased fruit and vegetable consumption, and increased stool astriction. The proportion of γδT cells was significantly higher in children with ASD than TD children (β: 0.156; 95% CI: 0.888 ∼ 2.135, p < 0.001) even after adjusting for gender, eating and mealtime behaviors, and dietary habits. In addition, the increased γδT cells were evident in all age groups (age < 48 months: β: 0.288; 95% CI: 0.420 ∼ 4.899, p = 0.020; age ≥ 48 months: β: 0.458; 95% CI: 0.694 ∼ 9.352, p = 0.024), as well as in boys (β: 0.174; 95% CI: 0.834 ∼ 2.625, p < 0.001) but not in girls. These findings were also confirmed by an external validation cohort. Furthermore, IL-17, but not IFN-γ, secretion by the circulating γδT cells was increased in ASD children. Machine learning revealed that the area under the curve in nomogram plots for increased γδT cells combined with eating behavior/dietary factors was 0.905, which held true in both boys and girls and in all the age groups of ASD children. The decision curves showed that children can receive significantly higher diagnostic benefit within the threshold probability range from 0 to 1.0 in the nomogram model. CONCLUSIONS Children with ASD present with divergent eating and mealtime behaviors and dietary habits as well as gastrointestinal symptoms. In peripheral blood, γδT cells but not αβT cells are associated with ASD. The increased γδT cells combined with eating and mealtime behavior/dietary factors have a high value for assisting in the diagnosis of ASD.
Collapse
Affiliation(s)
- Lingling Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yiran Xu
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Shuang Sun
- Center for Child Behavioral Development, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Cailing Liang
- Center for Child Behavioral Development, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Wenhua Li
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Hongwei Li
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xiaoli Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Dizhou Pang
- Center for Child Behavioral Development, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Mengyue Li
- Center for Child Behavioral Development, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Huihui Li
- Center for Child Behavioral Development, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yongbin Lang
- Center for Child Behavioral Development, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jiatian Liu
- Center for Child Behavioral Development, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Shuqin Jiang
- Center for Child Behavioral Development, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xiaoyi Shi
- Center for Child Behavioral Development, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Bingbing Li
- Center for Child Behavioral Development, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yanyan Yang
- Center for Child Behavioral Development, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yazhe Wang
- Center for Child Behavioral Development, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhenghua Li
- Center for Child Behavioral Development, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Chunlan Song
- Center for Child Behavioral Development, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Guiqin Duan
- Center for Child Behavioral Development, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jianmei W Leavenworth
- Department of Neurosurgery and Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Centre of Perinatal Medicine and Health, Institute of Clinical Science, University of Gothenburg, 40530 Gothenburg, Sweden.
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Göteborg 40530, Sweden.
| |
Collapse
|
40
|
Kamalmaz N, Ben Bacha A, Alonazi M, Albasher G, Khayyat AIA, El-Ansary A. Unveiling sex-based differences in developing propionic acid-induced features in mice as a rodent model of ASD. PeerJ 2023; 11:e15488. [PMID: 37334116 PMCID: PMC10274690 DOI: 10.7717/peerj.15488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 05/10/2023] [Indexed: 06/20/2023] Open
Abstract
Background Males are more likely to develop autism as a neurodevelopmental disorder than females are, although the mechanisms underlying male vulnerability are not fully understood. Therefore, studying the role of autism etiologies considering sex differences in the propionic acid (PPA) rodent model of autism would build greater understanding of how females are protected from autism spectrum disorder, which may be used as a treatment strategy for males with autism. Objectives This study aimed to investigate the sex differences in oxidative stress, glutamate excitotoxicity, neuroinflammation, and gut microbiota impairment as etiological mechanisms for many neurological diseases, with specific reference to autism. Method Forty albino mice were divided into four groups of 10 animals each with two control and two treated groups of both sexes received only phosphate-buffered saline or a neurotoxic dose of PPA (250 mg/kg body weight) for 3 days, respectively. Biochemical markers of energy metabolism, oxidative stress, neuroinflammation, and excitotoxicity were measured in mouse brain homogenates, whereas the presence of pathogenic bacteria was assessed in mouse stool samples. Furthermore, the repetitive behavior, cognitive ability, and physical-neural coordination of the animals were examined. Results Collectively, selected variables related to oxidative stress, glutamate excitotoxicity, neuroinflammation, and gut bacteria were impaired concomitantly with altered behavior in PPA-induced rodent model, with males being more susceptible than females. Conclusion This study explains the role of sex in the higher vulnerability of males to develop autistic biochemical and behavioral features compared with females. Female sex hormones and the higher detoxification capacity and higher glycolytic flux in females serve as neuroprotective contributors in a rodent model of autism.
Collapse
Affiliation(s)
- Nasreen Kamalmaz
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Abir Ben Bacha
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Mona Alonazi
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Gadah Albasher
- Zoology Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Arwa Ishaq A. Khayyat
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Afaf El-Ansary
- Central Research Laboratory, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
41
|
Kaminski VDL, Michita RT, Ellwanger JH, Veit TD, Schuch JB, Riesgo RDS, Roman T, Chies JAB. Exploring potential impacts of pregnancy-related maternal immune activation and extracellular vesicles on immune alterations observed in autism spectrum disorder. Heliyon 2023; 9:e15593. [PMID: 37305482 PMCID: PMC10256833 DOI: 10.1016/j.heliyon.2023.e15593] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 04/03/2023] [Accepted: 04/14/2023] [Indexed: 06/13/2023] Open
Abstract
Autism Spectrum Disorder (ASD) is a set of neurodevelopmental disorders usually observed in early life, with impacts on behavioral and social skills. Incidence of ASD has been dramatically increasing worldwide, possibly due to increase in awareness/diagnosis as well as to genetic and environmental triggers. Currently, it is estimated that ∼1% of the world population presents ASD symptoms. In addition to its genetic background, environmental and immune-related factors also influence the ASD etiology. In this context, maternal immune activation (MIA) has recently been suggested as a component potentially involved in ASD development. In addition, extracellular vesicles (EVs) are abundant at the maternal-fetal interface and are actively involved in the immunoregulation required for a healthy pregnancy. Considering that alterations in concentration and content of EVs have also been associated with ASD, this article raises a debate about the potential roles of EVs in the processes surrounding MIA. This represents the major differential of the present review compared to other ASD studies. To support the suggested correlations and hypotheses, findings regarding the roles of EVs during pregnancy and potential influences on ASD are discussed, along with a review and update concerning the participation of infections, cytokine unbalances, overweight and obesity, maternal anti-fetal brain antibodies, maternal fever, gestational diabetes, preeclampsia, labor type and microbiota unbalances in MIA and ASD.
Collapse
Affiliation(s)
- Valéria de Lima Kaminski
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul – UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
- Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul – UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
- Programa de Pós-Graduação em Biotecnologia, Laboratório de Imunologia Aplicada, Instituto de Ciência e Tecnologia - ICT, Universidade Federal de São Paulo - UNIFESP, São José dos Campos, São Paulo, Brazil
| | - Rafael Tomoya Michita
- Laboratório de Genética Molecular Humana, Universidade Luterana do Brasil - ULBRA, Canoas, Rio Grande do Sul, Brazil
| | - Joel Henrique Ellwanger
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul – UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
- Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul – UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Tiago Degani Veit
- Instituto de Ciências Básicas da Saúde, Departmento de Microbiologia, Imunologia e Parasitologia, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Jaqueline Bohrer Schuch
- Centro de Pesquisa em Álcool e Drogas, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
- Programa de Pós-Graduação em Psiquiatria e Ciências do Comportamento, Universidade Federal do Rio Grande do Sul – UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Rudimar dos Santos Riesgo
- Child Neurology Unit, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Tatiana Roman
- Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul – UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - José Artur Bogo Chies
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul – UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
- Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul – UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
42
|
Jiang P, Zhou L, Du Z, Zhao L, Tang Y, Fei X, Wang L, Li D, Li S, Yang H, Fan X, Liao H. Icariin alleviates autistic-like behavior, hippocampal inflammation and vGlut1 expression in adult BTBR mice. Behav Brain Res 2023; 445:114384. [PMID: 36889463 DOI: 10.1016/j.bbr.2023.114384] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/23/2023] [Accepted: 03/05/2023] [Indexed: 03/08/2023]
Abstract
Autism spectrum disorder (ASD) is a complicated, heterogeneous disorder characterized by social interaction deficits and repetitive stereotypical behaviors. Neuroinflammation and synaptic protein dysregulation have been implicated in ASD pathogenesis. Icariin (ICA) has proven to exert neuroprotective function through anti-inflammatory function. Therefore, this study aimed to clarify the effects of ICA treatment on autism-like behavioral deficits in BTBR mice and whether these changes were related to modifications in the hippocampal inflammation and the balance of excitatory/inhibitory synapses. ICA supplementation (80 mg/kg, once daily for ten days, i.g.) ameliorated social deficits, repetitive stereotypical behaviors, and short-term memory deficit without affecting locomotor activity or anxiety-like behaviors of BTBR mice. Furthermore, ICA treatment inhibited neuroinflammation via decreasing microglia number and the soma size in the CA1 region of the hippocampus, as well as the protein levels of proinflammatory cytokines in the hippocampus of BTBR mice. In addition, ICA treatment also rescued excitatory-inhibitory synaptic protein imbalance by inhibiting the increased vGlut1 level without affecting the vGAT level in the BTBR mouse hippocampus. Collectively, the observed results indicate that ICA treatment alleviates ASD-like features, mitigates disturbed balance of excitatory-inhibitory synaptic protein, and inhibits hippocampal inflammation in BTBR mice, and may represent a novel promising drug for ASD treatment.
Collapse
Affiliation(s)
- Peiyan Jiang
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China; Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing 400038, China
| | - Lianyu Zhou
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing 400038, China; Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Zhulin Du
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Linyang Zhao
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing 400038, China
| | - Yexi Tang
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Xinghang Fei
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing 400038, China; Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Lian Wang
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing 400038, China
| | - Dabing Li
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Song Li
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, China; Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, China
| | - Hui Yang
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, China; Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, China.
| | - Xiaotang Fan
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing 400038, China.
| | - Huiling Liao
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
43
|
Fucic A, Mantovani A, Vena J, Bloom MS, Sincic N, Vazquez M, Aguado-Sierra J. Impact of endocrine disruptors from mother's diet on immuno-hormonal orchestration of brain development and introduction of the virtual human twin tool. Reprod Toxicol 2023; 117:108357. [PMID: 36863570 DOI: 10.1016/j.reprotox.2023.108357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/19/2023] [Accepted: 02/22/2023] [Indexed: 03/04/2023]
Abstract
Diet has long been known to modify physiology during development and adulthood. However, due to a growing number of manufactured contaminants and additives over the last few decades, diet has increasingly become a source of exposure to chemicals that has been associated with adverse health risks. Sources of food contaminants include the environment, crops treated with agrochemicals, inappropriate storage (e.g., mycotoxins) and migration of xenobiotics from food packaging and food production equipment. Hence, consumers are exposed to a mixture of xenobiotics, some of which are endocrine disruptors (EDs). The complex interactions between immune function and brain development and their orchestration by steroid hormones are insufficiently understood in human populations, and little is known about the impact on immune-brain interactions by transplacental fetal exposure to EDs via maternal diet. To help to identify the key data gaps, this paper aims to present (a) how transplacental EDs modify immune system and brain development, and (b) how these mechanisms may correlate with diseases such as autism and disturbances of lateral brain development. Attention is given to disturbances of the subplate, a transient structure of crucial significance in brain development. Additionally, we describe cutting edge approaches to investigate the developmental neurotoxicity of EDs, such as the application of artificial intelligence and comprehensive modelling. In the future, highly complex investigations will be performed using virtual brain models constructed using sophisticated multi-physics/multi-scale modelling strategies based on patient and synthetic data, which will enable a greater understanding of healthy or disturbed brain development.
Collapse
Affiliation(s)
- A Fucic
- Institute for Medical Research and Occupational Health, Ksaverska C 2, Zagreb, Croatia.
| | - A Mantovani
- Istituto Superiore di Sanità, Department of Food Safety, Nutrition and Veterinary Public Health, Rome, Italy
| | - J Vena
- Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - M S Bloom
- Global and Community Health, George Mason University, 4400 University Dr., Fairfax, VA, USA
| | - N Sincic
- Medical School, University of Zagreb, Salata 3, Croatia
| | - M Vazquez
- Barcelona Supercomputing Center, Plaça Eusebi Güell, 1-3, Barcelona 08034, Spain
| | - J Aguado-Sierra
- Barcelona Supercomputing Center, Plaça Eusebi Güell, 1-3, Barcelona 08034, Spain
| |
Collapse
|
44
|
Influence of Immune System Abnormalities Caused by Maternal Immune Activation in the Postnatal Period. Cells 2023; 12:cells12050741. [PMID: 36899877 PMCID: PMC10001371 DOI: 10.3390/cells12050741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/15/2023] [Accepted: 02/21/2023] [Indexed: 03/03/2023] Open
Abstract
The developmental origins of health and disease (DOHaD) indicate that fetal tissues and organs in critical and sensitive periods of development are susceptible to structural and functional changes due to the adverse environment in utero. Maternal immune activation (MIA) is one of the phenomena in DOHaD. Exposure to maternal immune activation is a risk factor for neurodevelopmental disorders, psychosis, cardiovascular diseases, metabolic diseases, and human immune disorders. It has been associated with increased levels of proinflammatory cytokines transferred from mother to fetus in the prenatal period. Abnormal immunity induced by MIA includes immune overreaction or immune response failure in offspring. Immune overreaction is a hypersensitivity response of the immune system to pathogens or allergic factor. Immune response failure could not properly fight off various pathogens. The clinical features in offspring depend on the gestation period, inflammatory magnitude, inflammatory type of MIA in the prenatal period, and exposure to prenatal inflammatory stimulation, which might induce epigenetic modifications in the immune system. An analysis of epigenetic modifications caused by adverse intrauterine environments might allow clinicians to predict the onset of diseases and disorders before or after birth.
Collapse
|
45
|
Bhat RS, Alonazi M, Al-Daihan S, El-Ansary A. Prenatal SSRI Exposure Increases the Risk of Autism in Rodents via Aggravated Oxidative Stress and Neurochemical Changes in the Brain. Metabolites 2023; 13:metabo13020310. [PMID: 36837929 PMCID: PMC9963091 DOI: 10.3390/metabo13020310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
The mechanisms underlying selective serotonin reuptake inhibitor (SSRI) use during pregnancy as a major autism risk factor are unclear. Here, brain neurochemical changes following fluoxetine exposure and in an autism model were compared to determine the effects on autism risk. The study was performed on neonatal male western albino rats which were divided into Groups one (control), two (propionic acid [PPA]-induced autism model), and three (prenatal SSRI-exposed newborn rats whose mothers were exposed to 5 mg/kg of fluoxetine over gestation days 10-20). SSRI (fluoxetine) induced significant neurochemical abnormalities in the rat brain by increasing lipid peroxide (MDA), Interferon-gamma (IFN-γ), and caspase-3 levels and by depleting Glutathione (GSH), Glutathione S-transferases (GST), Catalase, potassium (K+), and Creatine kinase (CK) levels, similarly to what has been discovered in the PPA model of autism when compared with control. Prenatal fluoxetine exposure plays a significant role in asset brain damage in newborns; further investigation of fluoxetine as an autism risk factor is thus warranted.
Collapse
Affiliation(s)
- Ramesa Shafi Bhat
- Biochemistry Department, College of Science, King Saud University, Riyadh 11495, Saudi Arabia
- Correspondence:
| | - Mona Alonazi
- Biochemistry Department, College of Science, King Saud University, Riyadh 11495, Saudi Arabia
| | - Sooad Al-Daihan
- Biochemistry Department, College of Science, King Saud University, Riyadh 11495, Saudi Arabia
| | - Afaf El-Ansary
- Central Research Laboratory, Female Campus, King Saud University, Riyadh 11495, Saudi Arabia
| |
Collapse
|
46
|
Ashwood P. Preliminary Findings of Elevated Inflammatory Plasma Cytokines in Children with Autism Who Have Co-Morbid Gastrointestinal Symptoms. Biomedicines 2023; 11:436. [PMID: 36830973 PMCID: PMC9952966 DOI: 10.3390/biomedicines11020436] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Autism spectrum disorder (AU) is present in approximately 2% of the population and is often associated with co-morbidities that can impact quality of life. One of the most common co-morbidities in autism is the presence of gastrointestinal (GI) symptoms consisting of irregular bowel habits such as constipation, diarrhea, or alternating bowel habit. Evidence of immune infiltration and immune activation has been shown in the ileum and colon of children with AU with GI symptoms. Moreover, immune dysfunction is a contributing factor in many GI diseases, and we hypothesize that it would be more apparent in children with AU that exhibit GI symptoms than those who do not present with GI symptoms. The aim of this preliminary study was to determine whether there are altered cytokine levels in plasma in children with AU with GI symptoms compared with children with AU without GI symptoms, typically developing (TD) children with GI symptoms and TD children without GI symptoms, from the same population-based cohort. Plasma cytokine levels were assessed by multiplex assays. No differences in plasma cytokines were observed in TD controls with or without GI symptoms; however, many innate (IL-1α, TNFα, GM-CSF, IFNα) and adaptive cytokines (IL-4, IL-13, IL-12p70) were increased in AU children with GI symptoms compared with children with AU with no GI symptoms. The mucosal relevant cytokine IL-15 was increased in AU with GI symptoms compared with all groups. In contrast, the regulatory cytokine IL-10, was reduced in AU with GI symptoms and may suggest an imbalance in pro-inflammatory/regulatory signals. These data suggest that children with AU and GI symptoms have an imbalance in their immune response that is evident in their circulating plasma cytokine levels. A finding that could point to potential therapeutic and/or monitoring strategies for GI issues in AU.
Collapse
Affiliation(s)
- Paul Ashwood
- Department of Medical Microbiology and Immunology, School of Medicine, MIND Institute, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
47
|
Maurya SK, Gupta S, Mishra R. Transcriptional and epigenetic regulation of microglia in maintenance of brain homeostasis and neurodegeneration. Front Mol Neurosci 2023; 15:1072046. [PMID: 36698776 PMCID: PMC9870594 DOI: 10.3389/fnmol.2022.1072046] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/15/2022] [Indexed: 01/12/2023] Open
Abstract
The emerging role of microglia in brain homeostasis, neurodegeneration, and neurodevelopmental disorders has attracted considerable interest. In addition, recent developments in microglial functions and associated pathways have shed new light on their fundamental role in the immunological surveillance of the brain. Understanding the interconnections between microglia, neurons, and non-neuronal cells have opened up additional avenues for research in this evolving field. Furthermore, the study of microglia at the transcriptional and epigenetic levels has enhanced our knowledge of these native brain immune cells. Moreover, exploring various facets of microglia biology will facilitate the early detection, treatment, and management of neurological disorders. Consequently, the present review aimed to provide comprehensive insight on microglia biology and its influence on brain development, homeostasis, management of disease, and highlights microglia as potential therapeutic targets in neurodegenerative and neurodevelopmental diseases.
Collapse
Affiliation(s)
- Shashank Kumar Maurya
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, University of Delhi, New Delhi, India,*Correspondence: Shashank Kumar Maurya, ;
| | - Suchi Gupta
- Tech Cell Innovations Private Limited, Centre for Medical Innovation and Entrepreneurship (CMIE), All India Institute of Medical Sciences, New Delhi, India
| | - Rajnikant Mishra
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, India
| |
Collapse
|
48
|
Sun R, Guo P, Sun T, Yu H, Liao Y, Xie J, Zeng J, Xie X, Huang S. Sex hormone receptor expression in children with autism spectrum disorder.. [DOI: 10.21203/rs.3.rs-2345028/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Abstract
Background
Sex hormones, especially estrogen, which binds to estrogen receptor β (ERβ), play a vital role in the pathogenesis of mental disorders such as autism spectrum disorder (ASD). The purpose of this study was to analyze the serum levels of hormone receptors, including ERβ, progesterone receptor (PGR) and androgen receptor (AR), and compare these levels between children with ASD and typically developing (TD) children. We also investigated the relationships of ERβ mRNA levels with ASD core symptoms, sleep, and developmental quotients (DQs) from the Gesell Developmental Schedules (GDS) among children with ASD.
Methods
We compared the mRNA levels of ERβ, AR, and PGR between 56 children with ASD and 37 TD children by using quantitative real-time PCR. Then, a correlation analysis was performed to determine the correlations of ERβ mRNA levels with Childhood Autism Rating Scale (CARS), Autism Behavior Checklist (ABC), and Children’s Sleep Habits Questionnaire (CHSQ) scores as well as DQs among ASD children.
Results
We found that serum mRNA levels of ERβ in ASD children were significantly lower than those in the TD group. However, we found no correlations of the ERβ mRNA level with CARS, ABC, and CHSQ scores as well as DQs on each GDS domain among ASD children.
Conclusions
Elevated ERβ mRNA levels in peripheral blood may be related to ASD but this association needs to be validated with a larger sample size.
Collapse
Affiliation(s)
- Ruoyu Sun
- Foshan Maternal and Child Health Hospital, Southern Medical University
| | - Pi Guo
- Shantou University Medical College
| | - Tao Sun
- Foshan Maternal and Child Health Hospital, Southern Medical University
| | - Hong Yu
- Foshan Maternal and Child Health Hospital, Southern Medical University
| | - Yanwei Liao
- Foshan Maternal and Child Health Hospital, Southern Medical University
| | - Jieqi Xie
- Foshan Maternal and Child Health Hospital, Southern Medical University
| | - Jiaying Zeng
- Foshan Maternal and Child Health Hospital, Southern Medical University
| | - Xiaoyun Xie
- Foshan Maternal and Child Health Hospital, Southern Medical University
| | - Saijun Huang
- Foshan Maternal and Child Health Hospital, Southern Medical University
| |
Collapse
|
49
|
Eissa N, Awad MA, Thomas SD, Venkatachalam K, Jayaprakash P, Zhong S, Stark H, Sadek B. Simultaneous Antagonism at H3R/D2R/D3R Reduces Autism-like Self-Grooming and Aggressive Behaviors by Mitigating MAPK Activation in Mice. Int J Mol Sci 2022; 24:ijms24010526. [PMID: 36613969 PMCID: PMC9820264 DOI: 10.3390/ijms24010526] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/31/2022] Open
Abstract
Dysregulation in brain neurotransmitters underlies several neuropsychiatric disorders, e.g., autism spectrum disorder (ASD). Also, abnormalities in the extracellular-signal-regulated kinase (ERK)/mitogen-activated protein kinase (MAPK) pathway pave the way for neuroinflammation, neurodegeneration, and altered learning phenotype in ASD. Therefore, the effects of chronic systemic administration of the multiple-targeting antagonist ST-713 at the histamine H3 receptor (H3R) and dopamine D2/D3 receptors (D2/D3R) on repetitive self-grooming, aggressive behaviors, and abnormalities in the MAPK pathway in BTBR T + Itpr3tf/J (BTBR) mice were assessed. The results showed that ST-713 (2.5, 5, and 10 mg/kg, i.p.) mitigated repetitive self-grooming and aggression in BTBR mice (all p < 0.05), and the ameliorative effects of the most promising dose of ST-713 (5 mg/kg, i.p.) on behaviors were completely abrogated by co-administration of the H3R agonist (R)-α-methylhistamine or the anticholinergic drug scopolamine. Moreover, the elevated levels of several MAPK pathway proteins and induced proinflammatory markers such as tumor necrosis factor (TNF-α), interleukin-1β (IL-1β), and IL-6 were significantly suppressed following chronic administration of ST-713 (5 mg/kg, i.p.) (all p < 0.01). Furthermore, ST-713 significantly increased the levels of histamine and dopamine in hippocampal tissue of treated BTBR mice (all p < 0.01). The current observations signify the potential role of such multiple-targeting compounds, e.g., ST-713, in multifactorial neurodevelopmental disorders such as ASD.
Collapse
Affiliation(s)
- Nermin Eissa
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Zayed Bin Sultan Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi P.O. Box 59911, United Arab Emirates
| | - Mohamed Al Awad
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Zayed Bin Sultan Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Shilu Deepa Thomas
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Zayed Bin Sultan Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Karthikkumar Venkatachalam
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Zayed Bin Sultan Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Petrilla Jayaprakash
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Zayed Bin Sultan Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Sicheng Zhong
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitaetsstr. 1, 40225 Düsseldorf, Germany
| | - Holger Stark
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitaetsstr. 1, 40225 Düsseldorf, Germany
| | - Bassem Sadek
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Zayed Bin Sultan Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Correspondence:
| |
Collapse
|
50
|
Keil-Stietz K, Lein PJ. Gene×environment interactions in autism spectrum disorders. Curr Top Dev Biol 2022; 152:221-284. [PMID: 36707213 PMCID: PMC10496028 DOI: 10.1016/bs.ctdb.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
There is credible evidence that environmental factors influence individual risk and/or severity of autism spectrum disorders (hereafter referred to as autism). While it is likely that environmental chemicals contribute to the etiology of autism via multiple mechanisms, identifying specific environmental factors that confer risk for autism and understanding how they contribute to the etiology of autism has been challenging, in part because the influence of environmental chemicals likely varies depending on the genetic substrate of the exposed individual. Current research efforts are focused on elucidating the mechanisms by which environmental chemicals interact with autism genetic susceptibilities to adversely impact neurodevelopment. The goal is to not only generate insights regarding the pathophysiology of autism, but also inform the development of screening platforms to identify specific environmental factors and gene×environment (G×E) interactions that modify autism risk. Data from such studies are needed to support development of intervention strategies for mitigating the burden of this neurodevelopmental condition on individuals, their families and society. In this review, we discuss environmental chemicals identified as putative autism risk factors and proposed mechanisms by which G×E interactions influence autism risk and/or severity using polychlorinated biphenyls (PCBs) as an example.
Collapse
Affiliation(s)
- Kimberly Keil-Stietz
- Department of Comparative Biosciences, University of Wisconsin-Madison, School of Veterinary Medicine, Madison, WI, United States
| | - Pamela J Lein
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA, United States.
| |
Collapse
|