1
|
Tsurutani M, Goto T, Hagihara M, Irie S, Miyamichi K. Selective vulnerability of parvocellular oxytocin neurons in social dysfunction. Nat Commun 2024; 15:8661. [PMID: 39370447 PMCID: PMC11456597 DOI: 10.1038/s41467-024-53092-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 09/30/2024] [Indexed: 10/08/2024] Open
Abstract
Selective vulnerability offers a conceptual framework for understanding neurodegenerative disorders such as Parkinson's disease, where specific neuronal types are selectively affected and adjacent ones are spared. However, the applicability of this framework to neurodevelopmental disorders, particularly those characterized by atypical social behaviors, such as autism spectrum disorder, remains uncertain. Here we show that an embryonic disturbance, known to induce social dysfunction in male mice, preferentially impaired the gene expression crucial for neural functions in parvocellular oxytocin (OT) neurons-a subtype linked to social rewards-while neighboring cell types experienced a lesser impact. Chemogenetic stimulation of OT neurons at the neonatal stage ameliorated social deficits in early adulthood, concurrent with cell-type-specific sustained recovery of pivotal gene expression within parvocellular OT neurons. Collectively, our data shed light on the transcriptomic selective vulnerability within the hypothalamic social behavioral center and provide a potential therapeutic target through specific neonatal neurostimulation.
Collapse
Affiliation(s)
- Masafumi Tsurutani
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
- Graduate School of Biostudies, Kyoto University, Kyoto, Kyoto, Japan
| | - Teppei Goto
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Mitsue Hagihara
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Satsuki Irie
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Kazunari Miyamichi
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan.
| |
Collapse
|
2
|
Gan M, Zhu X, Wang W, Ye K, Jiang Y, Jiang T, Lv H, Lu Q, Qin R, Tao S, Huang L, Xu X, Liu C, Dou Y, Ke K, Sun T, Liu Y, Jiang Y, Han X, Jin G, Ma H, Shen H, Hu Z, Guan Y, Lin Y, Du J. Associations of inflammation related prenatal adversities with neurodevelopment of offspring in one year: a longitudinal prospective birth cohort study. BMC Pregnancy Childbirth 2024; 24:636. [PMID: 39358694 PMCID: PMC11445952 DOI: 10.1186/s12884-024-06839-8] [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: 03/20/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND The recent Maternal Immune Activation (MIA) theory suggests maternal systemic inflammation may serve as a mediator in associations between prenatal maternal adversities and neurodevelopmental diseases in offspring. Given the co-exposure to multiple adversities may be experienced by pregnant person, it is unclear whether a quantitative index can be developed to characterize the inflammation related exposure level, and whether this index is associated with neurodevelopmental delays in offspring. METHODS Based on Jiangsu Birth Cohort (JBC), a total of 3051 infants were included in the analysis. Inflammation related Prenatal Adversity Index (IPAI) was constructed using maternal data. Neurodevelopmental outcomes were assessed using the Bayley Scales of Infant and Toddler Development, third edition, screening test in one year. Multivariate linear regression and Poisson regression model were performed to analyze the associations between IPAI and neurodevelopment in offspring. RESULTS Compared with "low IPAI" group, offspring with "high IPAI" have lower scores of cognition, receptive communication, expressive communication, and fine motor. The adjusted β were - 0.23 (95%CI: -0.42, -0.04), -0.47 (95%CI: -0.66, -0.28), -0.30 (95%CI: -0.49, -0.11), and - 0.20 (95%CI: -0.33, -0.06). Additionally, the elevated risk for noncompetent development of cognition and receptive communication among "high IPAI" group was observed. The relative risk [RR] and 95% confidence interval [CI] were 1.35 (1.01, 1.69) and 1.37 (1.09, 1.72). CONCLUSIONS Our results revealed a significant association between higher IPAI and lower scores across cognition, receptive communication, expressive communication, and fine motor domains, and an increased risk of noncompetent development in the cognition and receptive communication domains.
Collapse
Affiliation(s)
- Ming Gan
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Taizhou People's Hospital, Affiliated to Nanjing Medical University, Taizhou, 225300, Jiangsu, China
| | - Xianxian Zhu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Maternal, Child and Adolescent Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Weiting Wang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Maternal, Child and Adolescent Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Kan Ye
- State Key Laboratory of Reproductive Medicine (Suzhou Centre), Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, 215002, Jiangsu, China
| | - Yangqian Jiang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Maternal, Child and Adolescent Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Tao Jiang
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Hong Lv
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- State Key Laboratory of Reproductive Medicine (Suzhou Centre), Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, 215002, Jiangsu, China
| | - Qun Lu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Maternal, Child and Adolescent Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Rui Qin
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Shiyao Tao
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Lei Huang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Maternal, Child and Adolescent Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Xin Xu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Maternal, Child and Adolescent Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Cong Liu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Yuanyan Dou
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Kang Ke
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Tianyu Sun
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Yuxin Liu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Yue Jiang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Xiumei Han
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Guangfu Jin
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Hongxia Ma
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- State Key Laboratory of Reproductive Medicine (Suzhou Centre), Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, 215002, Jiangsu, China
| | - Hongbing Shen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Zhibin Hu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- State Key Laboratory of Reproductive Medicine (Suzhou Centre), Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, 215002, Jiangsu, China
| | - Yichun Guan
- Department of Reproductive Medicine Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- State Key Laboratory of Reproductive Medicine (Henan Centre), The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Yuan Lin
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
- Department of Maternal, Child and Adolescent Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
- State Key Laboratory of Reproductive Medicine (Suzhou Centre), Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, 215002, Jiangsu, China.
- Department of Maternal, Child and Adolescent Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Jiangbo Du
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
- State Key Laboratory of Reproductive Medicine (Suzhou Centre), Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, 215002, Jiangsu, China.
- Taizhou People's Hospital, Affiliated to Nanjing Medical University, Taizhou, 225300, Jiangsu, China.
| |
Collapse
|
3
|
Baptista FI, Ambrósio AF. Tracing the influence of prenatal risk factors on the offspring retina: Focus on development and putative long-term consequences. Eur J Clin Invest 2024; 54:e14266. [PMID: 38864773 DOI: 10.1111/eci.14266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/10/2024] [Accepted: 05/29/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND Pregnancy represents a window of vulnerability to fetal development. Disruptions in the prenatal environment during this crucial period can increase the risk of the offspring developing diseases over the course of their lifetime. The central nervous system (CNS) has been shown to be particularly susceptible to changes during crucial developmental windows. To date, research focused on disruptions in the development of the CNS has predominantly centred on the brain, revealing a correlation between exposure to prenatal risk factors and the onset of neuropsychiatric disorders. Nevertheless, some studies indicate that the retina, which is part of the CNS, is also vulnerable to in utero alterations during pregnancy. Such changes may affect neuronal, glial and vascular components of the retina, compromising retinal structure and function and possibly impairing visual function. METHODS A search in the PubMed database was performed, and any literature concerning prenatal risk factors (drugs, diabetes, unbalanced diet, infection, glucocorticoids) affecting the offspring retina were included. RESULTS This review collects evidence on the cellular, structural and functional changes occurring in the retina triggered by maternal risk factors during pregnancy. We highlight the adverse impact on retinal development and its long-lasting effects, providing a critical analysis of the current knowledge while underlining areas for future research. CONCLUSIONS Appropriate recognition of the prenatal risk factors that negatively impact the developing retina may provide critical clues for the design of preventive strategies and for early therapeutic intervention that could change retinal pathology in the progeny.
Collapse
Affiliation(s)
- Filipa I Baptista
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - António F Ambrósio
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- Association for Innovation and Biomedical Research on Light and Image, Coimbra, Portugal
| |
Collapse
|
4
|
Cui S, Xiong W, Zhao Z, Han Y, Cui T, Qu Z, Li Z, Zhang X. Negative impact of maternal depressive symptoms on infancy neurodevelopment: a moderated mediation effect of maternal inflammation. Eur Child Adolesc Psychiatry 2024:10.1007/s00787-024-02572-x. [PMID: 39190153 DOI: 10.1007/s00787-024-02572-x] [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: 05/20/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024]
Abstract
Maternal depression promotes maternal inflammation and the risk of neurodevelopmental disorder in offspring, but the role of inflammation on the association between depression and neurodevelopment in offspring has not been extensively studied in humans. This study aims to examine the mediating role of maternal inflammation on the relationship between maternal depression and neurodevelopment in infants. 146 mother-child pairs were identified from Tianjin Maternal and Child Health Education and Service Cohort (Tianjin MCHESC). Maternal depression was investigated by the Center for Epidemiologic Studies Depression Scale and the Edinburgh Postnatal Depression Scale, and depressive trajectories were identified by latent class growth analysis. Inflammatory biomarkers in the three trimesters were assessed with enzyme-linked immunoassay. The Children Neuropsychological and Behavior Scale-Revision 2016 was used to measure neurodevelopment in infants. Principal component analysis was performed to identify inflammatory condition. Stepwise multiple linear regression analysis and mediation analysis were used to identify association among maternal depression, maternal inflammation and neurodevelopment in infants. Offspring in the low and moderate maternal depression groups exhibited higher adaptive behavior development quotient than those in the high maternal depression group. Higher maternal c-reactive protein level and higher inflammatory level in acute-phase of inflammation in the first trimester, and moderate maternal depression were associated with lower adaptive behavior quotients of infants. Inflammatory level in acute-phase of inflammation in the first trimester significantly mediated the association between maternal depression and adaptive behavior development of infants, with explaining 11.85% of the association. Maternal depression could impair adaptive behavior development in infants by inflammation.
Collapse
Affiliation(s)
- Shanshan Cui
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Wenjuan Xiong
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Ziyu Zhao
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Yu Han
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Tingkai Cui
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Zhiyi Qu
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Zhi Li
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Xin Zhang
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
5
|
Foo SS, Chen W, Azamor T, Jung KL, Cambou MC, Familiar-Macedo D, Salem GM, Melano I, Sim MS, Moreira ME, Brasil P, Vasconcelos Z, Nielsen-Saines K, Jung JU. Sustained chronic inflammation and altered childhood vaccine responses in children exposed to Zika virus. EBioMedicine 2024; 106:105249. [PMID: 39024898 PMCID: PMC11304698 DOI: 10.1016/j.ebiom.2024.105249] [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: 08/28/2023] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Congenital Zika virus (ZIKV) infection leads to severe newborn abnormalities, but its long-term impact on childhood immunity is not well understood. This study aims to investigate the serum proteomics in children exposed to ZIKV during pregnancy to understand potential immunological consequences during early childhood. METHODS The study included ZIKV-exposed infants (ZEI) at birth (n = 42) and children exposed to ZIKV (ZEC) at two years of age (n = 20) exposed to ZIKV during pregnancy, as well as healthy controls. Serum proteomic analysis was performed on these groups to assess inflammation and immune profiles. Additionally, antibody titres against two common childhood vaccines, DTaP and MMR, were measured in healthy controls (n = 50) and ZEC (n = 92) to evaluate vaccine-induced immunity. FINDINGS Results showed elevated inflammation in ZEI with birth abnormalities. Among ZEC, despite most having normal clinical outcomes at two years, their serum proteomics indicated a bias towards Th1-mediated immune responses. Notably, ZEC displayed reduced anti-Diphtheria toxin and anti-Clostridium tetani IgG levels against DTaP and MMR vaccines. They also exhibited lower antibody titres particularly against Th2-biased DTaP vaccines, but not Th1-biased MMR vaccines. INTERPRETATION In conclusion, the study highlights the long-term immunological consequences of congenital ZIKV exposure. Heightened inflammation was observed in ZEI with abnormalities at birth, while ZEC maintained a chronic Th1-biased immune profile. The impaired response to Th2-biased vaccines raises concerns about lasting effects of ZIKV exposure on immune responses. Consequently, there is a need for continued longitudinal clinical monitoring to identify potential immune-related complications arising from prenatal exposure to ZIKV. FUNDING This work was partially funded by the National Institute of Allergy and Infectious Diseases (NIAID) and National Institute of Dental and Craniofacial Research (NIDCR).
Collapse
Affiliation(s)
- Suan-Sin Foo
- Department of Infection Biology and Global Centre for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| | - Weiqiang Chen
- Department of Infection Biology and Global Centre for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Tamiris Azamor
- Department of Infection Biology and Global Centre for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Kyle L Jung
- Department of Infection Biology and Global Centre for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Mary Catherine Cambou
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Débora Familiar-Macedo
- Department of Infection Biology and Global Centre for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Gielenny M Salem
- Department of Infection Biology and Global Centre for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ivonne Melano
- Department of Infection Biology and Global Centre for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Myung-Shin Sim
- Department of Medicine, Statistics Core, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | | | | | - Zilton Vasconcelos
- Department of Medicine, Statistics Core, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Karin Nielsen-Saines
- Fundação Oswaldo Cruz, Rio de Janeiro, Brazil; Department of Paediatrics, Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jae U Jung
- Department of Infection Biology and Global Centre for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
6
|
Tu G, Jiang N, Chen W, Liu L, Hu M, Liao B. The neurobiological mechanisms underlying the effects of exercise interventions in autistic individuals. Rev Neurosci 2024; 0:revneuro-2024-0058. [PMID: 39083671 DOI: 10.1515/revneuro-2024-0058] [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: 04/22/2024] [Accepted: 07/15/2024] [Indexed: 08/02/2024]
Abstract
Autism spectrum disorder is a pervasive and heterogeneous neurodevelopmental condition characterized by social communication difficulties and rigid, repetitive behaviors. Owing to the complex pathogenesis of autism, effective drugs for treating its core features are lacking. Nonpharmacological approaches, including education, social-communication, behavioral and psychological methods, and exercise interventions, play important roles in supporting the needs of autistic individuals. The advantages of exercise intervention, such as its low cost, easy implementation, and high acceptance, have garnered increasing attention. Exercise interventions can effectively improve the core features and co-occurring conditions of autism, but the underlying neurobiological mechanisms are unclear. Abnormal changes in the gut microbiome, neuroinflammation, neurogenesis, and synaptic plasticity may individually or interactively be responsible for atypical brain structure and connectivity, leading to specific autistic experiences and characteristics. Interestingly, exercise can affect these biological processes and reshape brain network connections, which may explain how exercise alleviates core features and co-occurring conditions in autistic individuals. In this review, we describe the definition, diagnostic approach, epidemiology, and current support strategies for autism; highlight the benefits of exercise interventions; and call for individualized programs for different subtypes of autistic individuals. Finally, the possible neurobiological mechanisms by which exercise improves autistic features are comprehensively summarized to inform the development of optimal exercise interventions and specific targets to meet the needs of autistic individuals.
Collapse
Affiliation(s)
- Genghong Tu
- Department of Sports Medicine, 47878 Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, 47878 Scientific Research Center, Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
| | - Nan Jiang
- Graduate School, 47878 Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
| | - Weizhong Chen
- Graduate School, 47878 Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
| | - Lining Liu
- Graduate School, 47878 Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
| | - Min Hu
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, 47878 Scientific Research Center, Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
| | - Bagen Liao
- Department of Sports Medicine, 47878 Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, 47878 Scientific Research Center, Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
| |
Collapse
|
7
|
Li S, Zhang N, Li W, Zhang HL, Wang XX. Gastrointestinal problems in a valproic acid-induced rat model of autism: From maternal intestinal health to offspring intestinal function. World J Psychiatry 2024; 14:1095-1105. [PMID: 39050201 PMCID: PMC11262932 DOI: 10.5498/wjp.v14.i7.1095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/13/2024] [Accepted: 06/04/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a developmental disorder characterized by social deficits and repetitive behavior. Gastrointestinal (GI) problems, such as constipation, diarrhea, and inflammatory bowel disease, commonly occur in patients with ASD. Previously, GI problems of ASD patients were attributed to intestinal inflammation and vertical mother-to-infant microbiome transmission. AIM To explore whether GI problems in ASD are related to maternal intestinal inflammation and gut microbiota abnormalities. METHODS An ASD rat model was developed using valproic acid (VPA). Enzyme-linked immunosorbent assay and fecal 16S rRNA sequencing were used to test GI changes. RESULTS VPA exposure during pregnancy led to pathological maternal intestinal changes, resulting in alterations in maternal gut microbiota. Additionally, the levels of inflammatory factors also increased. Moreover, prenatal exposure to VPA resulted in impaired duodenal motility in the offspring as well as increased levels of inflammatory factors. CONCLUSION GI problems in ASD may be associated with maternal intestinal inflammation and microbiota abnormality. Future research is required to find more evidence on the etiology and treatment of GI problems in ASD.
Collapse
Affiliation(s)
- Sha Li
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing 100000, China
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100000, China
| | - Nan Zhang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing 100000, China
| | - Wang Li
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100000, China
| | - Han-Lai Zhang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing 100000, China
| | - Xiao-Xi Wang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing 100000, China
| |
Collapse
|
8
|
Pike MR, Lipner E, O'Brien KJ, Breen EC, Cohn BA, Cirillo PM, Krigbaum NY, Kring AM, Olino TM, Alloy LB, Ellman LM. Prenatal maternal Inflammation, childhood cognition and adolescent depressive symptoms. Brain Behav Immun 2024; 119:908-918. [PMID: 38761818 DOI: 10.1016/j.bbi.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 04/10/2024] [Accepted: 05/12/2024] [Indexed: 05/20/2024] Open
Abstract
BACKGROUND Accumulating evidence indicates that higher prenatal maternal inflammation is associated with increased depression risk in adolescent and adult-aged offspring. Prenatal maternal inflammation (PNMI) may increase the likelihood for offspring to have lower cognitive performance, which, in turn, may heighten risk for depression onset. Therefore, this study explored the potential mediating role of childhood cognitive performance in the relationship between PNMI and adolescent depressive symptoms in offspring. METHODS Participants included 696 mother-offspring dyads from the Child Health and Development Studies (CHDS) cohort. Biomarkers of maternal inflammation [interleukin (IL)-6, IL-8, IL-1 receptor antagonist (IL-1RA) and soluble TNF receptor-II (sTNF-RII)] were assayed from first (T1) and second trimester (T2) sera. Childhood (ages 9-11) cognitive performance was assessed via standardized Peabody Picture Vocabulary Test (PPVT), a measure of receptive vocabulary correlated with general intelligence. Adolescent (ages 15-17) depressive symptoms were assessed via self-report. RESULTS There were no significant associations between T1 biomarkers and childhood PPVT or adolescent depressive symptoms. Higher T2 IL1-RA was directly associated with lower childhood PPVT (b = -0.21, SE = 0.08, t = -2.55, p = 0.01), but not with adolescent depressive symptoms. T2 IL-6 was not directly associated with childhood PPVT, but higher T2 IL-6 was directly associated at borderline significance with greater depressive symptoms in adolescence (b = 0.05, SE = 0.03, t = 1.96, p = 0.05). Lower childhood PPVT predicted significantly higher adolescent depressive symptoms (b = -0.07, SE = 0.02, t = -2.99, p < 0.01). There was a significant indirect effect of T2 IL-1RA on adolescent depressive symptoms via childhood PPVT (b = 0.03, 95 % CI = 0.002-0.03) indicating a partially mediated effect. No significant associations were found with T2 sTNF-RII nor IL-8. CONCLUSIONS Lower childhood cognitive performance, such as that indicated by a lower PPVT score, represents a potential mechanism through which prenatal maternal inflammation contributes to adolescent depression risk in offspring.
Collapse
Affiliation(s)
- Madeline R Pike
- Temple University, Department of Psychology and Neuroscience, 1701 N 13th St, Philadelphia, PA 19122, USA.
| | - Emily Lipner
- Temple University, Department of Psychology and Neuroscience, 1701 N 13th St, Philadelphia, PA 19122, USA
| | - Kathleen J O'Brien
- Temple University, Department of Psychology and Neuroscience, 1701 N 13th St, Philadelphia, PA 19122, USA
| | - Elizabeth C Breen
- Cousins Center for Psychoneuroimmunology, University of California-Los Angeles, 300 Medical Plaza, Suite 3306, Los Angeles, CA 90095-7076, USA
| | - Barbara A Cohn
- Child Health and Development Studies, Public Health Institute, 1683 Shattuck Ave., Suite B, Berkeley, CA 94709, USA
| | - Piera M Cirillo
- Child Health and Development Studies, Public Health Institute, 1683 Shattuck Ave., Suite B, Berkeley, CA 94709, USA
| | - Nickilou Y Krigbaum
- Child Health and Development Studies, Public Health Institute, 1683 Shattuck Ave., Suite B, Berkeley, CA 94709, USA
| | - Ann M Kring
- University of California, Berkeley, Department of Psychology, 2121 Berkeley Way, Berkeley, CA 94720, USA
| | - Thomas M Olino
- Temple University, Department of Psychology and Neuroscience, 1701 N 13th St, Philadelphia, PA 19122, USA
| | - Lauren B Alloy
- Temple University, Department of Psychology and Neuroscience, 1701 N 13th St, Philadelphia, PA 19122, USA
| | - Lauren M Ellman
- Temple University, Department of Psychology and Neuroscience, 1701 N 13th St, Philadelphia, PA 19122, USA
| |
Collapse
|
9
|
Carey ME, Kivumbi A, Rando J, Mesaros AC, Melnyk S, James SJ, Croen LA, Volk H, Lyall K. The association between prenatal oxidative stress levels measured by isoprostanes and offspring neurodevelopmental outcomes at 36 months. Brain Behav Immun Health 2024; 38:100775. [PMID: 38706573 PMCID: PMC11067487 DOI: 10.1016/j.bbih.2024.100775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 04/16/2024] [Indexed: 05/07/2024] Open
Abstract
Oxidative stress during pregnancy has been a mechanistic pathway implicated in autism development, yet few studies have examined this association directly. Here, we examined the association of prenatal levels of 8-iso-PGF2α, a widely used measure of oxidative stress, and several neurodevelopmental outcomes related to autism in children. Participants included 169 mother-child pairs from the Early Autism Risk Longitudinal Investigation (EARLI), which enrolled mothers who had an autistic child from a previous pregnancy and followed them through a subsequent pregnancy and until that child reached age 3 years. Maternal urine samples were collected during the second trimester of pregnancy and were later measured for levels of isoprostanes. Child neurodevelopmental assessments included the Mullen Scales of Early Learning (MSEL), the Social Responsiveness Scale (SRS), and the Vineland Adaptive Behavior Scale (VABS), and were conducted around 36 months of age. Primary analyses examined associations between interquartile range (IQR) increases in 8-iso-PGF2α levels, and total composite scores from each assessment using quantile regression. In adjusted analyses, we did not observe statistically significant associations, though estimates suggested modestly lower cognitive scores (β for MSEL = -3.68, 95% CI: -10.09, 2.70), and minor increases in autism-related trait scores (β for SRS T score = 1.68, 95% CI: -0.24, 3.60) with increasing 8-iso-PGF2α. These suggestive associations between decreased cognitive scores and increased autism-related traits with increasing prenatal oxidative stress point to the need for continued investigation in larger samples of the role of oxidative stress as a mechanistic pathway in autism and related neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Meghan E. Carey
- A.J. Drexel Autism Institute, Drexel University, 3020 Market Street, Suite 560, Philadelphia, PA, 19104, USA
| | - Apollo Kivumbi
- A.J. Drexel Autism Institute, Drexel University, 3020 Market Street, Suite 560, Philadelphia, PA, 19104, USA
| | - Juliette Rando
- A.J. Drexel Autism Institute, Drexel University, 3020 Market Street, Suite 560, Philadelphia, PA, 19104, USA
| | - A. Clementina Mesaros
- Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA, 17104, USA
| | - Stepan Melnyk
- Arkansas Children’s Hospital Research Institute, 13 Childrens Way, Little Rock, AR, 72202, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, 4301 W Markham St, Little Rock, AR, 72205, USA
| | - S. Jill James
- Department of Pediatrics, University of Arkansas for Medical Sciences, 4301 W Markham St, Little Rock, AR, 72205, USA
| | - Lisa A. Croen
- Division of Research Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA, 94612, USA
| | - Heather Volk
- Department of Mental Health, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe St, Baltimore, MD, 21205, USA
| | - Kristen Lyall
- A.J. Drexel Autism Institute, Drexel University, 3020 Market Street, Suite 560, Philadelphia, PA, 19104, USA
| | - the Early Autism Risk Longitudinal Investigation (EARLI) team
- A.J. Drexel Autism Institute, Drexel University, 3020 Market Street, Suite 560, Philadelphia, PA, 19104, USA
- Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA, 17104, USA
- Arkansas Children’s Hospital Research Institute, 13 Childrens Way, Little Rock, AR, 72202, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, 4301 W Markham St, Little Rock, AR, 72205, USA
- Division of Research Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA, 94612, USA
- Department of Mental Health, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe St, Baltimore, MD, 21205, USA
| |
Collapse
|
10
|
Shen J, Liu L, Yang Y, Zhou M, Xu S, Zhang W, Zhang C. Insulin-Like Growth Factor 1 Has the Potential to Be Used as a Diagnostic Tool and Treatment Target for Autism Spectrum Disorders. Cureus 2024; 16:e65393. [PMID: 39188438 PMCID: PMC11346671 DOI: 10.7759/cureus.65393] [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] [Accepted: 07/16/2024] [Indexed: 08/28/2024] Open
Abstract
Autism spectrum disorder (ASD), a heterogeneous group of neurodevelopmental disorders, is characterized by social impairment and repetitive and stereotypic behaviors. Because of the lack of approved laboratory diagnostic markers and effective therapeutic medications, it is one of the most challenging diseases. Therefore, it is urgent to explore potential diagnosis markers or therapeutic targets. Insulin-like growth factor 1 (IGF-1) is a neurotrophic growth factor that enhances brain development. IGF-1 levels in body fluids are lower in preschool children with ASD than in typically developing children, which may serve as a potential diagnostic marker. In various ASD models associated with genetic or environmental exposure, IGF-1 treatment can improve core symptoms or pathological changes, including neuronal development, neural cell survival, balance of synaptic excitation and inhibition, neuroimmunology, and oxidative stress status. In March 2023 an IGF-1 derivative was approved as the first drug for treating Rett syndrome, an ASD-related neurodevelopmental disorder, to improve fundamental symptoms such as social communication. Thus, in this review, we present accumulating evidence of altered IGF-1 levels in ASD patients and the possible mechanisms, as well as evidence that IGF-1 treatment improves the pathophysiology in various ASD models. IGF-1 has the potential to be an early diagnosis marker and an effective therapeutic for ASD.
Collapse
Affiliation(s)
- Jiamin Shen
- Department of Children Health Care, Jingmen Maternity and Child Health Care Hospital, Jingmen, CHN
| | - Lijuan Liu
- Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, CHN
| | - Yifan Yang
- Department of Children Health Care, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital) Tongji Medical College, Huazhong University of Science and Technology, Wuhan, CHN
| | - Miao Zhou
- Department of Children Health Care, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital) Tongji Medical College, Huazhong University of Science and Technology, Wuhan, CHN
| | - Shan Xu
- Department of Children Health Care, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital) Tongji Medical College, Huazhong University of Science and Technology, Wuhan, CHN
| | - Wanqing Zhang
- Department of Children Health Care, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital) Tongji Medical College, Huazhong University of Science and Technology, Wuhan, CHN
| | - Chuanjie Zhang
- Department of Children Health Care, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital) Tongji Medical College, Huazhong University of Science and Technology, Wuhan, CHN
| |
Collapse
|
11
|
Zhang W, Mou Z, Zhong Q, Liu X, Yan L, Gou L, Chen Z, So KF, Zhang L. Transcutaneous auricular vagus nerve stimulation improves social deficits through the inhibition of IL-17a signaling in a mouse model of autism. Front Psychiatry 2024; 15:1393549. [PMID: 38993386 PMCID: PMC11237520 DOI: 10.3389/fpsyt.2024.1393549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/11/2024] [Indexed: 07/13/2024] Open
Abstract
Background Maternal exposure to inflammation is one of the causes of autism spectrum disorder (ASD). Electrical stimulation of the vagus nerve exerts a neuroprotective effect via its anti-inflammatory action. We thus investigated whether transcutaneous auricular vagus nerve stimulation (taVNS) can enhance social abilities in a mouse model of ASD induced by maternal immune activation (MIA). Methods ASD mouse model were constructed by intraperitoneal injection of polyinosinic:polycytidylic acid (poly (I:C)). TaVNS with different parameters were tested in ASD mouse model and in C57BL/6 mice, then various behavioral tests and biochemical analyses related to autism were conducted. ASD model mice were injected with an interleukin (IL)-17a antibody into the brain, followed by behavioral testing and biochemical analyses. Results TaVNS reduced anxiety, improved social function, decreased the number of microglia, and inhibited M1 polarization of microglia. Additionally, taVNS attenuated the expression of the IL-17a protein in the prefrontal cortex and blood of ASD model mice. To examine the possible involvement of IL-17a in taVNS-induced neuroprotection, we injected an IL-17a antibody into the prefrontal cortex of ASD model mice and found that neutralizing IL-17a decreased the number of microglia and inhibited M1 polarization. Furthermore, neutralizing IL-17a improved social function in autism model mice. Conclusion Our study revealed that reduced neuroinflammation is an important mechanism of taVNS-mediated social improvement and neuroprotection against autism. This effect of taVNS could be attributed to the inhibition of the IL-17a pathway.
Collapse
Affiliation(s)
- Wenjing Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zhiwei Mou
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Department of Rehabilitation Medicine, The Fifth Affiliated Hospital of Jinan University, Heyuan, China
| | - Qi Zhong
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiaocao Liu
- Lab of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Lan Yan
- Key Laboratory of Central Nervous System (CNS) Regeneration (Ministry of Education), Guangdong–Hong Kong–Macau Institute of Central Nervous System (CNS) Regeneration, Jinan University, Guangzhou, China
| | - Lei Gou
- Department of Rehabilitation Medicine, The Fifth Affiliated Hospital of Jinan University, Heyuan, China
| | - Zhuoming Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Kwok-Fai So
- Key Laboratory of Central Nervous System (CNS) Regeneration (Ministry of Education), Guangdong–Hong Kong–Macau Institute of Central Nervous System (CNS) Regeneration, Jinan University, Guangzhou, China
| | - Li Zhang
- Key Laboratory of Central Nervous System (CNS) Regeneration (Ministry of Education), Guangdong–Hong Kong–Macau Institute of Central Nervous System (CNS) Regeneration, Jinan University, Guangzhou, China
| |
Collapse
|
12
|
Jyonouchi H. Autism spectrum disorder and a possible role of anti-inflammatory treatments: experience in the pediatric allergy/immunology clinic. Front Psychiatry 2024; 15:1333717. [PMID: 38979496 PMCID: PMC11228311 DOI: 10.3389/fpsyt.2024.1333717] [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: 11/05/2023] [Accepted: 06/03/2024] [Indexed: 07/10/2024] Open
Abstract
Autism spectrum disorder (ASD1) is a behaviorally defined syndrome encompassing a markedly heterogeneous patient population. Many ASD subjects fail to respond to the 1st line behavioral and pharmacological interventions, leaving parents to seek out other treatment options. Evidence supports that neuroinflammation plays a role in ASD pathogenesis. However, the underlying mechanisms likely vary for each ASD patient, influenced by genetic, epigenetic, and environmental factors. Although anti-inflammatory treatment measures, mainly based on metabolic changes and oxidative stress, have provided promising results in some ASD subjects, the use of such measures requires the careful selection of ASD subjects based on clinical and laboratory findings. Recent progress in neuroscience and molecular immunology has made it possible to allow re-purposing of currently available anti-inflammatory medications, used for autoimmune and other chronic inflammatory conditions, as treatment options for ASD subjects. On the other hand, emerging anti-inflammatory medications, including biologic and gate-keeper blockers, exert powerful anti-inflammatory effects on specific mediators or signaling pathways. It will require both a keen understanding of the mechanisms of action of such agents and the careful selection of ASD patients suitable for each treatment. This review will attempt to summarize the use of anti-inflammatory agents already used in targeting ASD patients, and then emerging anti-inflammatory measures applicable for ASD subjects based on scientific rationale and clinical trial data, if available. In our experience, some ASD patients were treated under diagnoses of autoimmune/autoinflammatory conditions and/or post-infectious neuroinflammation. However, there are little clinical trial data specifically for ASD subjects. Therefore, these emerging immunomodulating agents for potential use for ASD subjects will be discussed based on preclinical data, case reports, or data generated in patients with other medical conditions. This review will hopefully highlight the expanding scope of immunomodulating agents for treating neuroinflammation in ASD subjects.
Collapse
Affiliation(s)
- Harumi Jyonouchi
- Department of Pediatrics, Saint Peter's University Hospital, New Brunswick, NJ, United States
- Department of Pediatrics, Rutgers University-Robert Wood Johnson School of Medicine, New Brunswick, NJ, United States
| |
Collapse
|
13
|
Wikarska A, Roszak K, Roszek K. Mesenchymal Stem Cells and Purinergic Signaling in Autism Spectrum Disorder: Bridging the Gap between Cell-Based Strategies and Neuro-Immune Modulation. Biomedicines 2024; 12:1310. [PMID: 38927517 PMCID: PMC11201695 DOI: 10.3390/biomedicines12061310] [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: 04/28/2024] [Revised: 05/26/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
The prevalence of autism spectrum disorder (ASD) is still increasing, which means that this neurodevelopmental lifelong pathology requires special scientific attention and efforts focused on developing novel therapeutic approaches. It has become increasingly evident that neuroinflammation and dysregulation of neuro-immune cross-talk are specific hallmarks of ASD, offering the possibility to treat these disorders by factors modulating neuro-immunological interactions. Mesenchymal stem cell-based therapy has already been postulated as one of the therapeutic approaches for ASD; however, less is known about the molecular mechanisms of stem cell influence. One of the possibilities, although still underestimated, is the paracrine purinergic activity of MSCs, by which stem cells ameliorate inflammatory reactions. Modulation of adenosine signaling may help restore neurotransmitter balance, reduce neuroinflammation, and improve overall brain function in individuals with ASD. In our review article, we present a novel insight into purinergic signaling, including but not limited to the adenosinergic pathway and its role in neuroinflammation and neuro-immune cross-talk modulation. We anticipate that by achieving a greater understanding of the purinergic signaling contribution to ASD and related disorders, novel therapeutic strategies may be devised for patients with autism in the near future.
Collapse
Affiliation(s)
| | | | - Katarzyna Roszek
- Department of Biochemistry, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Torun, Lwowska 1, 87-100 Torun, Poland; (A.W.); (K.R.)
| |
Collapse
|
14
|
Ionescu MI, Zahiu CDM, Vlad A, Galos F, Gradisteanu Pircalabioru G, Zagrean AM, O'Mahony SM. Nurturing development: how a mother's nutrition shapes offspring's brain through the gut. Nutr Neurosci 2024:1-23. [PMID: 38781488 DOI: 10.1080/1028415x.2024.2349336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Pregnancy is a transformative period marked by profound physical and emotional changes, with far-reaching consequences for both mother and child. Emerging research has illustrated the pivotal role of a mother's diet during pregnancy in influencing the prenatal gut microbiome and subsequently shaping the neurodevelopment of her offspring. The intricate interplay between maternal gut health, nutrition, and neurodevelopmental outcomes has emerged as a captivating field of investigation within developmental science. Acting as a dynamic bridge between mother and fetus, the maternal gut microbiome, directly and indirectly, impacts the offspring's neurodevelopment through diverse pathways. This comprehensive review delves into a spectrum of studies, clarifying putative mechanisms through which maternal nutrition, by modulating the gut microbiota, orchestrates the early stages of brain development. Drawing insights from animal models and human cohorts, this work underscores the profound implications of maternal gut health for neurodevelopmental trajectories and offers a glimpse into the formulation of targeted interventions able to optimize the health of both mother and offspring. The prospect of tailored dietary recommendations for expectant mothers emerges as a promising and accessible intervention to foster the growth of beneficial gut bacteria, potentially leading to enhanced cognitive outcomes and reduced risks of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Mara Ioana Ionescu
- Department of Functional Sciences, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Department of Pediatrics, Marie Curie Emergency Children's Hospital, Bucharest, Romania
| | - Carmen Denise Mihaela Zahiu
- Department of Functional Sciences, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Adelina Vlad
- Department of Functional Sciences, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Felicia Galos
- Department of Pediatrics, Marie Curie Emergency Children's Hospital, Bucharest, Romania
- Department of Pediatrics, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Gratiela Gradisteanu Pircalabioru
- Research Institute of the University of Bucharest, Section Earth, Environmental and Life Sciences, Section-ICUB, Bucharest, Romania
- Academy of Romanian Scientists, Bucharest, Romania
| | - Ana-Maria Zagrean
- Department of Functional Sciences, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Siobhain M O'Mahony
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
15
|
Xie X, Zhou R, Fang Z, Zhang Y, Wang Q, Liu X. Seeing beyond words: Visualizing autism spectrum disorder biomarker insights. Heliyon 2024; 10:e30420. [PMID: 38694128 PMCID: PMC11061761 DOI: 10.1016/j.heliyon.2024.e30420] [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: 10/07/2023] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/04/2024] Open
Abstract
Objective This study employs bibliometric and visual analysis to elucidate global research trends in Autism Spectrum Disorder (ASD) biomarkers, identify critical research focal points, and discuss the potential integration of diverse biomarker modalities for precise ASD assessment. Methods A comprehensive bibliometric analysis was conducted using data from the Web of Science Core Collection database until December 31, 2022. Visualization tools, including R, VOSviewer, CiteSpace, and gCLUTO, were utilized to examine collaborative networks, co-citation patterns, and keyword associations among countries, institutions, authors, journals, documents, and keywords. Results ASD biomarker research emerged in 2004, accumulating a corpus of 4348 documents by December 31, 2022. The United States, with 1574 publications and an H-index of 213, emerged as the most prolific and influential country. The University of California, Davis, contributed significantly with 346 publications and an H-index of 69, making it the leading institution. Concerning journals, the Journal of Autism and Developmental Disorders, Autism Research, and PLOS ONE were the top three publishers of ASD biomarker-related articles among a total of 1140 academic journals. Co-citation and keyword analyses revealed research hotspots in genetics, imaging, oxidative stress, neuroinflammation, gut microbiota, and eye tracking. Emerging topics included "DNA methylation," "eye tracking," "metabolomics," and "resting-state fMRI." Conclusion The field of ASD biomarker research is dynamically evolving. Future endeavors should prioritize individual stratification, methodological standardization, the harmonious integration of biomarker modalities, and longitudinal studies to advance the precision of ASD diagnosis and treatment.
Collapse
Affiliation(s)
- Xinyue Xie
- The First Affiliated Hospital of Henan University of Chinese Medicine, Pediatrics Hospital, Zhengzhou, Henan, 450000, China
- Henan University of Chinese Medicine, School of Pediatrics, Zhengzhou, Henan, 450046, China
| | - Rongyi Zhou
- The First Affiliated Hospital of Henan University of Chinese Medicine, Pediatrics Hospital, Zhengzhou, Henan, 450000, China
- Henan University of Chinese Medicine, School of Pediatrics, Zhengzhou, Henan, 450046, China
| | - Zihan Fang
- Henan University of Chinese Medicine, School of Pediatrics, Zhengzhou, Henan, 450046, China
| | - Yongting Zhang
- The First Affiliated Hospital of Henan University of Chinese Medicine, Pediatrics Hospital, Zhengzhou, Henan, 450000, China
- Henan University of Chinese Medicine, School of Pediatrics, Zhengzhou, Henan, 450046, China
| | - Qirong Wang
- Henan University of Chinese Medicine, School of Pediatrics, Zhengzhou, Henan, 450046, China
| | - Xiaomian Liu
- Henan University of Chinese Medicine, School of Medicine, Zhengzhou, Henan, 450046, China
| |
Collapse
|
16
|
Jain A, Hakim S, Woolf CJ. Immune drivers of physiological and pathological pain. J Exp Med 2024; 221:e20221687. [PMID: 38607420 PMCID: PMC11010323 DOI: 10.1084/jem.20221687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 09/25/2023] [Accepted: 04/02/2024] [Indexed: 04/13/2024] Open
Abstract
Physiological pain serves as a warning of exposure to danger and prompts us to withdraw from noxious stimuli to prevent tissue damage. Pain can also alert us of an infection or organ dysfunction and aids in locating such malfunction. However, there are instances where pain is purely pathological, such as unresolved pain following an inflammation or injury to the nervous system, and this can be debilitating and persistent. We now appreciate that immune cells are integral to both physiological and pathological pain, and that pain, in consequence, is not strictly a neuronal phenomenon. Here, we discuss recent findings on how immune cells in the skin, nerve, dorsal root ganglia, and spinal cord interact with somatosensory neurons to mediate pain. We also discuss how both innate and adaptive immune cells, by releasing various ligands and mediators, contribute to the initiation, modulation, persistence, or resolution of various modalities of pain. Finally, we propose that the neuroimmune axis is an attractive target for pain treatment, but the challenges in objectively quantifying pain preclinically, variable sex differences in pain presentation, as well as adverse outcomes associated with immune system modulation, all need to be considered in the development of immunotherapies against pain.
Collapse
Affiliation(s)
- Aakanksha Jain
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA, USA
| | - Sara Hakim
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Clifford J. Woolf
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Guo T, Zeng Z, Lin L. Exploration of the impact of dysbiosis in the gut microbiota on microbial composition in children's neurodevelopment. Int J Neurosci 2024:1-7. [PMID: 38606533 DOI: 10.1080/00207454.2024.2341924] [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: 02/24/2024] [Accepted: 04/07/2024] [Indexed: 04/13/2024]
Abstract
OBJECTIVE To investigate the impact of gut microbiota dysbiosis on neurodevelopment in children. METHODS This study included 338 children aged 0-3 years admitted to our hospital from January to December 2022, The children were divided into a normal neurodevelopment group (169 cases) and a poor neurodevelopment group (169 cases). Basic personal information and clinical data were collected through a detailed questionnaire, and the microbial composition in fecal samples was analyzed using 16S rRNA gene sequencing. RESULTS Children in the poor neurodevelopment group showed a significant decrease in gut microbiota diversity compared to those in the normal neurodevelopment group (Shannon index, p < 0.05). The abundance of Bifidobacterium and Veillonella genera significantly decreased (p < 0.05), while the abundance of Streptococcus genus increased significantly (p < 0.05). CONCLUSION There is an association between gut microbiota dysbiosis and poor neurodevelopment in children. The increased abundance of Streptococcus genus and decreased abundance of Bifidobacterium and Veillonella genera in the gut microbiota may be potential risk factors for poor neurodevelopment in preterm infants. Future research should further explore the potential beneficial effects of gut microbiota modulation on neurodevelopment in children.
Collapse
Affiliation(s)
- Ting Guo
- Department of Pediatrics, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Zhenzhong Zeng
- Department of Pediatrics, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Liangfeng Lin
- Department of Pediatrics, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| |
Collapse
|
18
|
Abad C, Karahoda R, Orbisova A, Kastner P, Heblik D, Kucera R, Portillo R, Staud F. Pathological shifts in tryptophan metabolism in human term placenta exposed to LPS or poly I:C†. Biol Reprod 2024; 110:722-738. [PMID: 38145492 PMCID: PMC11017130 DOI: 10.1093/biolre/ioad181] [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: 08/16/2023] [Revised: 10/25/2023] [Accepted: 12/18/2023] [Indexed: 12/27/2023] Open
Abstract
Maternal immune activation during pregnancy is a risk factor for offspring neuropsychiatric disorders. Among the mechanistic pathways by which maternal inflammation can affect fetal brain development and programming, those involving tryptophan (TRP) metabolism have drawn attention because various TRP metabolites have neuroactive properties. This study evaluates the effect of bacterial (lipopolysaccharides/LPS) and viral (polyinosinic:polycytidylic acid/poly I:C) placental infection on TRP metabolism using an ex vivo model. Human placenta explants were exposed to LPS or poly I:C, and the release of TRP metabolites was analyzed together with the expression of related genes and proteins and the functional activity of key enzymes in TRP metabolism. The rate-limiting enzyme in the serotonin pathway, tryptophan hydroxylase, showed reduced expression and functional activity in explants exposed to LPS or poly I:C. Conversely, the rate-limiting enzyme in the kynurenine pathway, indoleamine dioxygenase, exhibited increased activity, gene, and protein expression, suggesting that placental infection mainly promotes TRP metabolism via the kynurenine (KYN) pathway. Furthermore, we observed that treatment with LPS or poly I:C increased activity in the kynurenine monooxygenase branch of the KYN pathway. We conclude that placental infection impairs TRP homeostasis, resulting in decreased production of serotonin and an imbalance in the ratio between quinolinic acid and kynurenic acid. This disrupted homeostasis may eventually expose the fetus to suboptimal/toxic levels of neuroactive molecules and impair fetal brain development.
Collapse
Affiliation(s)
- Cilia Abad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Rona Karahoda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Anna Orbisova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Petr Kastner
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Daniel Heblik
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Radim Kucera
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Ramon Portillo
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Frantisek Staud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| |
Collapse
|
19
|
Fortin O, DeBiasi RL, Mulkey SB. Congenital infectious encephalopathies from the intrapartum period to postnatal life. Semin Fetal Neonatal Med 2024:101526. [PMID: 38677956 DOI: 10.1016/j.siny.2024.101526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2024]
Abstract
Congenital infections are a common but often underrecognized cause of fetal brain abnormalities, as well as fetal-neonatal morbidity and mortality, that should be considered by all healthcare professionals providing neurological care to fetuses and newborns. Maternal infection with various pathogens (cytomegalovirus, Toxoplasmosis, Rubella virus, Parvovirus B19, lymphocytic choriomeningitis virus, syphilis, Zika virus, varicella zoster virus) during pregnancy can be transmitted to the developing fetus, which can cause multisystem dysfunction and destructive or malformative central nervous system lesions. These can be recognized on fetal and neonatal imaging, including ultrasound and MRI. Imaging and clinical features often overlap, but some distinguishing features can help identify specific pathogens and guide subsequent testing strategies. Some pathogens can be specifically treated, and others can be managed with targeted interventions or symptomatic therapy based on expected complications. Neurological and neurodevelopmental complications related to congenital infections vary widely and are likely driven by a combination of pathophysiologic factors, alone or in combination. These include direct invasion of the fetal central nervous system by pathogens, inflammation of the maternal-placental-fetal triad in response to infection, and long-term effects of immunogenic and epigenetic changes in the fetus in response to maternal-fetal infection. Congenital infections and their neurodevelopmental impacts should be seen as an issue of public health policy, given that infection and the associated complications disproportionately affect woman and children from low- and middle-income countries and those with lower socio-economic status in high-income countries. Congenital infections may be preventable and treatable, which can improve long-term neurodevelopmental outcomes in children.
Collapse
Affiliation(s)
- Olivier Fortin
- Zickler Family Prenatal Pediatrics Institute, Children's National Hospital, Washington DC, USA
| | - Roberta L DeBiasi
- Division of Pediatric Infectious Disease, Children's National Hospital, Washington DC, USA; Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington DC, USA; Department of Tropical Medicine, Microbiology and Infectious Diseases, The George Washington University School of Medicine and Health Sciences, Washington DC, USA
| | - Sarah B Mulkey
- Zickler Family Prenatal Pediatrics Institute, Children's National Hospital, Washington DC, USA; Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington DC, USA; Department of Neurology and Rehabilitation Medicine, The George Washington University School of Medicine and Health Sciences, Washington DC, USA.
| |
Collapse
|
20
|
Graf S, Schechter DS. The Impact of Maternal Interpersonal Violent Trauma and Related Psychopathology on Child Outcomes and Intergenerational Transmission. Curr Psychiatry Rep 2024; 26:166-175. [PMID: 38427205 PMCID: PMC10978628 DOI: 10.1007/s11920-024-01491-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/26/2024] [Indexed: 03/02/2024]
Abstract
PURPOSE OF REVIEW This review aims to outline some consequences that maternal history of trauma with and without related psychopathology, such as posttraumatic stress symptoms (PTSS), can have on their children's development and functioning. It then addresses mechanisms through which intergenerational transmission of interpersonal violence (IPV) and related psychopathology may occur. RECENT FINDINGS Findings include the effects of maternal IPV experience and related psychopathology on child social-emotional and biologically-based outcomes. This includes increased developmental disturbances and child psychopathology, as well as physiological factors. Secondly, the review focuses on psychobiological mechanisms by which maternal experience of IPV and related psychopathology likely trigger intergenerational effects. Maternal IPV and related psychopathology can have a negative impact on several areas of their child's life including development, interactive behavior, psychopathology, and physiology. This transmission may partially be due to fetal and perinatal processes, genetic and epigenetic effects, and interactions with their parents.
Collapse
Affiliation(s)
- Shannen Graf
- Division of Child and Adolescent Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), SUPEA-Unité de recherche, Avenue d'Echallens 9, 1004, Lausanne, Switzerland.
- Department of Psychiatry, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.
| | - Daniel S Schechter
- Division of Child and Adolescent Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), SUPEA-Unité de recherche, Avenue d'Echallens 9, 1004, Lausanne, Switzerland
- Department of Psychiatry, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Department of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
21
|
Kim E, Huh JR, Choi GB. Prenatal and postnatal neuroimmune interactions in neurodevelopmental disorders. Nat Immunol 2024; 25:598-606. [PMID: 38565970 DOI: 10.1038/s41590-024-01797-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 02/15/2024] [Indexed: 04/04/2024]
Abstract
The intricate relationship between immune dysregulation and neurodevelopmental disorders (NDDs) has been observed across the stages of both prenatal and postnatal development. In this Review, we provide a comprehensive overview of various maternal immune conditions, ranging from infections to chronic inflammatory conditions, that impact the neurodevelopment of the fetus during pregnancy. Furthermore, we examine the presence of immunological phenotypes, such as immune-related markers and coexisting immunological disorders, in individuals with NDDs. By delving into these findings, we shed light on the potential underlying mechanisms responsible for the high occurrence of immune dysregulation alongside NDDs. We also discuss current mouse models of NDDs and their contributions to our understanding of the immune mechanisms underlying these diseases. Additionally, we discuss how neuroimmune interactions contribute to shaping the manifestation of neurological phenotypes in individuals with NDDs while also exploring potential avenues for mitigating these effects.
Collapse
Affiliation(s)
- Eunha Kim
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea.
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea.
| | - Jun R Huh
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Gloria B Choi
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
22
|
Jenkins HM, Meeker JD, Zimmerman E, Cathey A, Fernandez J, Montañez GH, Park S, Pabón ZR, Vélez Vega CM, Cordero JF, Alshawabkeh A, Watkins DJ. Gestational glyphosate exposure and early childhood neurodevelopment in a Puerto Rico birth cohort. ENVIRONMENTAL RESEARCH 2024; 246:118114. [PMID: 38211716 DOI: 10.1016/j.envres.2024.118114] [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: 11/16/2023] [Revised: 12/28/2023] [Accepted: 01/03/2024] [Indexed: 01/13/2024]
Abstract
INTRODUCTION N-(phosphonomethyl)glycine, or glyphosate, is a non-selective systemic herbicide widely used in agricultural, industrial, and residential settings since 1974. Glyphosate exposure has been inconsistently linked to neurotoxicity in animals, and studies of effects of gestational exposure among humans are scarce. In this study we investigated relationships between prenatal urinary glyphosate analytes and early childhood neurodevelopment. METHODS Mother-child pairs from the PROTECT-CRECE birth cohort in Puerto Rico with measures for both maternal urinary glyphosate analytes and child neurodevelopment were included for analysis (n = 143). Spot urine samples were collected 1-3 times throughout pregnancy and analyzed for glyphosate and aminomethylphosphonic acid (AMPA), an environmental degradant of glyphosate. Child neurodevelopment was assessed at 6, 12, and 24 months using the Battelle Developmental Inventory, 2nd edition Spanish (BDI-2), which provides scores for adaptive, personal-social, communication, motor, and cognitive domains. We used multivariable linear regression to examine associations between the geometric mean of maternal urinary glyphosate analytes across pregnancy and BDI-2 scores at each follow-up. Results were expressed as percent change in BDI-2 score per interquartile range increase in exposure. RESULTS Prenatal AMPA concentrations were negatively associated with communication domain at 12 months (%change = -5.32; 95%CI: 9.04, -1.61; p = 0.007), and communication subdomain scores at 12 and 24 months. At 24 months, four BDI-2 domains were associated with AMPA: adaptive (%change = -3.15; 95%CI: 6.05, -0.25; p = 0.038), personal-social (%change = -4.37; 95%CI: 7.48, -1.26; p = 0.008), communication (%change = -7.00; 95%CI: 11.75, -2.26; p = 0.005), and cognitive (%change = -4.02; 95%CI: 6.72, -1.32; p = 0.005). Similar trends were observed with GLY concentrations, but most confidence intervals include zero. We found no significant associations at 6 months. CONCLUSIONS Our results suggest that gestational exposure to glyphosate is associated with adverse early neurodevelopment, with more pronounced delays at 24 months. Given glyphosate's wide usage, further investigation into the impact of gestational glyphosate exposure on neurodevelopment is warranted.
Collapse
Affiliation(s)
- Haley M Jenkins
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA.
| | - John D Meeker
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA.
| | - Emily Zimmerman
- Department of Communication Sciences and Disorders, Northeastern University, Boston, MA, 02115, USA.
| | - Amber Cathey
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA.
| | - Jennifer Fernandez
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA.
| | - Gredia Huerta Montañez
- Department of Electrical and Computer Engineering, Northeastern University, Boston, MA, 02115, USA.
| | - Seonyoung Park
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA.
| | - Zaira Rosario Pabón
- Department of Electrical and Computer Engineering, Northeastern University, Boston, MA, 02115, USA.
| | - Carmen M Vélez Vega
- Department of Social Sciences, UPR Medical Sciences Campus, University of Puerto Rico Graduate School of Public Health, San Juan, PR, 00936, USA.
| | - José F Cordero
- Department of Epidemiology and Biostatistics, University of Georgia, Athens, GA, 30602, USA.
| | - Akram Alshawabkeh
- Department of Civil and Environmental Engineering, Northeastern University, Boston, MA, 02115, USA.
| | - Deborah J Watkins
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
23
|
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
|
24
|
Liu J, Liu JB, Ke XY. [Research progress on the mechanism of the impact of maternal childhood trauma on intergenerational transmission]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:207-212. [PMID: 38436321 PMCID: PMC10921875 DOI: 10.7499/j.issn.1008-8830.2309147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/05/2024] [Indexed: 03/05/2024]
Abstract
Childhood trauma refers to trauma experiences encountered during childhood and adolescence. Maternal childhood trauma experiences have a lasting impact on the next generation, affecting their physical and mental well-being. The mechanisms involved include the hypothalamic-pituitary-adrenal axis, inflammatory factors, brain structure and function, gene interactions, and parenting styles. This paper systematically reviews the mechanisms of the impact of maternal childhood trauma on intergenerational transmission, providing insights for the prevention of intergenerational transmission of childhood trauma.
Collapse
Affiliation(s)
- Juan Liu
- School of Mental Health, Jining Medical University, Jining, Shandong 272000, China (Ke X-Y, ); Department of Child Psychiatry, Shenzhen Mental Health Center/Shenzhen Kangning Hospital, Shenzhen, Guangdong 518000, China (Liu J-B, 308017398@qq. com)
| | | | - Xiao-Yin Ke
- School of Mental Health, Jining Medical University, Jining, Shandong 272000, China (Ke X-Y, ); Department of Child Psychiatry, Shenzhen Mental Health Center/Shenzhen Kangning Hospital, Shenzhen, Guangdong 518000, China (Liu J-B, 308017398@qq. com)
| |
Collapse
|
25
|
Sanders AFP, Tirado B, Seider NA, Triplett RL, Lean RE, Neil JJ, Miller JP, Tillman R, Smyser TA, Barch DM, Luby JL, Rogers CE, Smyser CD, Warner BB, Chen E, Miller GE. Prenatal exposure to maternal disadvantage-related inflammatory biomarkers: associations with neonatal white matter microstructure. Transl Psychiatry 2024; 14:72. [PMID: 38307841 PMCID: PMC10837200 DOI: 10.1038/s41398-024-02782-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 02/04/2024] Open
Abstract
Prenatal exposure to heightened maternal inflammation has been associated with adverse neurodevelopmental outcomes, including atypical brain maturation and psychiatric illness. In mothers experiencing socioeconomic disadvantage, immune activation can be a product of the chronic stress inherent to such environmental hardship. While growing preclinical and clinical evidence has shown links between altered neonatal brain development and increased inflammatory states in utero, the potential mechanism by which socioeconomic disadvantage differentially impacts neural-immune crosstalk remains unclear. In the current study, we investigated associations between socioeconomic disadvantage, gestational inflammation, and neonatal white matter microstructure in 320 mother-infant dyads over-sampled for poverty. We analyzed maternal serum levels of four cytokines (IL-6, IL-8, IL-10, TNF-α) over the course of pregnancy in relation to offspring white matter microstructure and socioeconomic disadvantage. Higher average maternal IL-6 was associated with very low socioeconomic status (SES; INR < 200% poverty line) and lower neonatal corticospinal fractional anisotropy (FA) and lower uncinate axial diffusivity (AD). No other cytokine was associated with SES. Higher average maternal IL-10 was associated with lower FA and higher radial diffusivity (RD) in corpus callosum and corticospinal tracts, higher optic radiation RD, lower uncinate AD, and lower FA in inferior fronto-occipital fasciculus and anterior limb of internal capsule tracts. SES moderated the relationship between average maternal TNF-α levels during gestation and neonatal white matter diffusivity. When these interactions were decomposed, the patterns indicated that this association was significant and positive among very low SES neonates, whereby TNF-α was inversely and significantly associated with inferior cingulum AD. By contrast, among the more advantaged neonates (lower-to-higher SES [INR ≥ 200% poverty line]), TNF-α was positively and significantly associated with superior cingulum AD. Taken together, these findings suggest that the relationship between prenatal cytokine exposure and white matter microstructure differs as a function of SES. These patterns are consistent with a scenario where gestational inflammation's effects on white matter development diverge depending on the availability of foundational resources in utero.
Collapse
Affiliation(s)
- Ashley F P Sanders
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - Brian Tirado
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Nicole A Seider
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Regina L Triplett
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Rachel E Lean
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jeffrey J Neil
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - J Philip Miller
- Division of Biostatistics, Institute for Informatics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Rebecca Tillman
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Tara A Smyser
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Deanna M Barch
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Psychological and Brain Sciences, Washington University School of Medicine, St. Louis, MO, 63130, USA
| | - Joan L Luby
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Cynthia E Rogers
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Christopher D Smyser
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Barbara B Warner
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Newborn Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Edith Chen
- Institute for Policy Research, Northwestern University, Evanston, IL, 60208, USA
- Department of Psychology, Northwestern University, Evanston, IL, 60208, USA
| | - Gregory E Miller
- Institute for Policy Research, Northwestern University, Evanston, IL, 60208, USA
- Department of Psychology, Northwestern University, Evanston, IL, 60208, USA
| |
Collapse
|
26
|
Huang ZY, Lyu ZP, Li HG, You HZ, Yang XN, Cha CH. Des-Arg(9) bradykinin as a causal metabolite for autism spectrum disorder. World J Psychiatry 2024; 14:88-101. [PMID: 38327885 PMCID: PMC10845217 DOI: 10.5498/wjp.v14.i1.88] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/08/2023] [Accepted: 12/07/2023] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND Early diagnosis and therapeutic interventions can greatly enhance the developmental trajectory of children with autism spectrum disorder (ASD). However, the etiology of ASD is not completely understood. The presence of confounding factors from environment and genetics has increased the difficulty of the identification of diagnostic biomarkers for ASD. AIM To estimate and interpret the causal relationship between ASD and metabolite profile, taking into consideration both genetic and environmental influences. METHODS A two-sample Mendelian randomization (MR) analysis was conducted using summarized data from large-scale genome-wide association studies (GWAS) including a metabolite GWAS dataset covering 453 metabolites from 7824 European and an ASD GWAS dataset comprising 18381 ASD cases and 27969 healthy controls. Metabolites in plasma were set as exposures with ASD as the main outcome. The causal relationships were estimated using the inverse variant weight (IVW) algorithm. We also performed leave-one-out sensitivity tests to validate the robustness of the results. Based on the drafted metabolites, enrichment analysis was conducted to interpret the association via constructing a protein-protein interaction network with multi-scale evidence from databases including Infinome, SwissTargetPrediction, STRING, and Metascape. RESULTS Des-Arg(9)-bradykinin was identified as a causal metabolite that increases the risk of ASD (β = 0.262, SE = 0.064, PIVW = 4.64 × 10-5). The association was robust, with no significant heterogeneity among instrument variables (PMR Egger = 0.663, PIVW = 0.906) and no evidence of pleiotropy (P = 0.949). Neuroinflammation and the response to stimulus were suggested as potential biological processes mediating the association between Des-Arg(9) bradykinin and ASD. CONCLUSION Through the application of MR, this study provides practical insights into the potential causal association between plasma metabolites and ASD. These findings offer perspectives for the discovery of diagnostic or predictive biomarkers to support clinical practice in treating ASD.
Collapse
Affiliation(s)
- Zhong-Yu Huang
- Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou 510000, Guangdong Province, China
| | - Zi-Pan Lyu
- School of Biological Sciences, Nanyang Technological University, Nanyang Ave 639798, Singapore
| | - Hong-Gui Li
- Department of Pediatrics, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong Province, China
| | - Hua-Zhi You
- Department of Nutrition, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong Province, China
| | - Xiang-Na Yang
- Department of Pediatric Traditional Chinese Medicine Clinic, Guangzhou Women and Children's Medical Center, Guangzhou 510632, Guangdong Province, China
| | - Cai-Hui Cha
- Department of Psychology, Guangzhou Women and Children's Medical Center, Guangzhou 510632, Guangdong Province, China
| |
Collapse
|
27
|
Manti S, Spoto G, Nicotera AG, Di Rosa G, Piedimonte G. Impact of respiratory viral infections during pregnancy on the neurological outcomes of the newborn: current knowledge. Front Neurosci 2024; 17:1320319. [PMID: 38260010 PMCID: PMC10800711 DOI: 10.3389/fnins.2023.1320319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/14/2023] [Indexed: 01/24/2024] Open
Abstract
Brain development is a complex process that begins during pregnancy, and the events occurring during this sensitive period can affect the offspring's neurodevelopmental outcomes. Respiratory viral infections are frequently reported in pregnant women, and, in the last few decades, they have been related to numerous neuropsychiatric sequelae. Respiratory viruses can disrupt brain development by directly invading the fetal circulation through vertical transmission or inducing neuroinflammation through the maternal immune activation and production of inflammatory cytokines. Influenza virus gestational infection has been consistently associated with psychotic disorders, such as schizophrenia and autism spectrum disorder, while the recent pandemic raised some concerns regarding the effects of severe acute respiratory syndrome coronavirus 2 on neurodevelopmental outcomes of children born to affected mothers. In addition, emerging evidence supports the possible role of respiratory syncytial virus infection as a risk factor for adverse neuropsychiatric consequences. Understanding the mechanisms underlying developmental dysfunction allows for improving preventive strategies, early diagnosis, and prompt interventions.
Collapse
Affiliation(s)
- Sara Manti
- Pediatric Unit, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, Messina, Italy
| | - Giulia Spoto
- Unit of Child Neurology and Psychiatry, Department of Biomedical and Dental Sciences and of Morphological and Functional Imaging, University of Messina, Messina, Italy
| | - Antonio Gennaro Nicotera
- Unit of Child Neurology and Psychiatry, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, Messina, Italy
| | - Gabriella Di Rosa
- Unit of Child Neurology and Psychiatry, Department of Biomedical and Dental Sciences and of Morphological and Functional Imaging, University of Messina, Messina, Italy
| | - Giovanni Piedimonte
- Department of Pediatrics, Biochemistry and Molecular Biology, Tulane University, New Orleans, LA, United States
| |
Collapse
|
28
|
Abarca-Castro EA, Talavera-Peña AK, Reyes-Lagos JJ, Becerril-Villanueva E, Pérez-Sanchez G, de la Peña FR, Maldonado-García JL, Pavón L. Modulation of vagal activity may help reduce neurodevelopmental damage in the offspring of mothers with pre-eclampsia. Front Immunol 2023; 14:1280334. [PMID: 38022681 PMCID: PMC10653300 DOI: 10.3389/fimmu.2023.1280334] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Maternal Immune Activation (MIA) has been linked to the pathogenesis of pre-eclampsia and adverse neurodevelopmental outcomes in the offspring, such as cognitive deficits, behavioral abnormalities, and mental disorders. Pre-eclampsia is associated with an activation of the immune system characterized by persistently elevated levels of proinflammatory cytokines, as well as a decrease in immunoregulatory factors. The Cholinergic Anti-inflammatory Pathway (CAP) may play a relevant role in regulating the maternal inflammatory response during pre-eclampsia and protecting the developing fetus from inflammation-induced damage. Dysregulation in the CAP has been associated with the clinical evolution of pre-eclampsia. Some studies suggest that therapeutic stimulation of this pathway may improve maternal and fetal outcomes in preclinical models of pre-eclampsia. Modulation of vagal activity influences the CAP, improving maternal hemodynamics, limiting the inflammatory response, and promoting the growth of new neurons, which enhances synaptic plasticity and improves fetal neurodevelopment. Therefore, we postulate that modulation of vagal activity may improve maternal and fetal outcomes in pre-eclampsia by targeting underlying immune dysregulation and promoting better fetal neurodevelopment. In this perspective, we explore the clinical and experimental evidence of electrical, pharmacological, physical, and biological stimulation mechanisms capable of inducing therapeutical CAP, which may be applied in pre-eclampsia to improve the mother's and offspring's quality of life.
Collapse
Affiliation(s)
- Eric Alonso Abarca-Castro
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Lerma (UAM-L), Lerma, Mexico
| | - Ana Karen Talavera-Peña
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Lerma (UAM-L), Lerma, Mexico
| | - José Javier Reyes-Lagos
- Facultad de Medicina, Universidad Autónoma del Estado de México (UAEMéx), Toluca de Lerdo, Mexico
| | - Enrique Becerril-Villanueva
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Gilberto Pérez-Sanchez
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Francisco R. de la Peña
- Unidad de Fomento a la Investigación, Dirección de Servicios Clínicos, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - José Luis Maldonado-García
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| |
Collapse
|
29
|
Sawada K, Kamiya S, Kobayashi T. Neonatal Exposure to Lipopolysaccharide Promotes Neurogenesis of Subventricular Zone Progenitors in the Developing Neocortex of Ferrets. Int J Mol Sci 2023; 24:14962. [PMID: 37834410 PMCID: PMC10573966 DOI: 10.3390/ijms241914962] [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: 09/06/2023] [Revised: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023] Open
Abstract
Lipopolysaccharide (LPS) is a natural agonist of toll-like receptor 4 that serves a role in innate immunity. The current study evaluated the LPS-mediated regulation of neurogenesis in the subventricular zone (SVZ) progenitors, that is, the basal radial glia and intermediate progenitors (IPs), in ferrets. Ferret pups were subcutaneously injected with LPS (500 μg/g of body weight) on postnatal days (PDs) 6 and 7. Furthermore, 5-ethynyl-2'-deoxyuridine (EdU) and 5-bromo-2'-deoxyuridine (BrdU) were administered on PDs 5 and 7, respectively, to label the post-proliferative and proliferating cells in the inner SVZ (iSVZ) and outer SVZ (oSVZ). A significantly higher density of BrdU single-labeled proliferating cells was observed in the iSVZ of LPS-exposed ferrets than in controls but not in post-proliferative EdU single-labeled and EdU/BrdU double-labeled self-renewing cells. BrdU single-labeled cells exhibited a lower proportion of Tbr2 immunostaining in LPS-exposed ferrets (22.2%) than in controls (42.6%) and a higher proportion of Ctip2 immunostaining in LPS-exposed ferrets (22.2%) than in controls (8.6%). The present findings revealed that LPS modified the neurogenesis of SVZ progenitors. Neonatal LPS exposure facilitates the proliferation of SVZ progenitors, followed by the differentiation of Tbr2-expressing IPs into Ctip2-expressing immature neurons.
Collapse
Affiliation(s)
- Kazuhiko Sawada
- Department of Nutrition, Faculty of Medical and Health Sciences, Tsukuba International University, Tsuchiura 300-0051, Japan
| | - Shiori Kamiya
- Department of Regulation Biology, Faculty of Science, Saitama University, Saitama 338-8570, Japan; (S.K.); (T.K.)
| | - Tetsuya Kobayashi
- Department of Regulation Biology, Faculty of Science, Saitama University, Saitama 338-8570, Japan; (S.K.); (T.K.)
| |
Collapse
|
30
|
Wu D, Li Y, Chen L, Klein M, Franke B, Chen J, Buitelaar J. Maternal gestational weight gain and offspring's neurodevelopmental outcomes: A systematic review and meta-analysis. Neurosci Biobehav Rev 2023; 153:105360. [PMID: 37573899 DOI: 10.1016/j.neubiorev.2023.105360] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 08/04/2023] [Accepted: 08/10/2023] [Indexed: 08/15/2023]
Abstract
Abnormal gestational weight gain (GWG) has been increasing globally, up to 47% of all pregnancies. Multiple studies have focused on the association between GWG and adverse neurodevelopmental outcomes in the offspring, however with inconsistent results. We performed a systematic review and meta-analysis to evaluate associations between excessive or insufficient GWG and offspring's neurodevelopmental outcomes. Meta-analysis of these 23 studies using a random-effects model revealed associations between excessive GWG and neurodevelopmental disorders (ASD & ID & ADHD together: OR=1.12 [95% CI 1.06-1.19]), ASD (OR=1.18 [95% CI 1.08-1.29]), ADHD (OR=1.08 [95% CI 1.02-1.14]), ASD with ID (OR=1.15 [95% CI 1.01-1.32]), and ASD without ID (OR=1.12 [95% CI 1.06-1.19]). Insufficient GWG was associated with higher risk for ID (OR=1.14 [95% CI 1.03-1.26]). These results emphasize the significant impact, though of small effect size, of GWG across multiple neurodevelopmental disorders. It is important to note that these results do not establish causality. Other factors such as genetic factors, gene-environment interactions may confound the relationship between GWG and neurodevelopmental outcomes. To better understand the role of GWG in neurodevelopmental disorders, future studies should consider using genetically sensitive designs that can account for these potential confounders.
Collapse
Affiliation(s)
- Dan Wu
- Department of Child Health Care, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200062, Shanghai, China; Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands; Department of Cognitive Neuroscience, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Yicheng Li
- Department of Child Health Care, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200062, Shanghai, China
| | - Lingyan Chen
- Department of Occupational Therapy Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki-city, 852-8520, Japan
| | - Marieke Klein
- Department of Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Barbara Franke
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands; Department of Cognitive Neuroscience, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands; Department of Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands; Department of Psychiatry, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Jinjin Chen
- Department of Child Health Care, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200062, Shanghai, China.
| | - Jan Buitelaar
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands; Department of Cognitive Neuroscience, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands; Karakter Child and Adolescent Psychiatry University Centre, 6525 GA Nijmegen, the Netherlands
| |
Collapse
|
31
|
Woods R, Lorusso J, Fletcher J, ElTaher H, McEwan F, Harris I, Kowash H, D'Souza SW, Harte M, Hager R, Glazier JD. Maternal immune activation and role of placenta in the prenatal programming of neurodevelopmental disorders. Neuronal Signal 2023; 7:NS20220064. [PMID: 37332846 PMCID: PMC10273029 DOI: 10.1042/ns20220064] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023] Open
Abstract
Maternal infection during pregnancy, leading to maternal immune activation (mIA) and cytokine release, increases the offspring risk of developing a variety of neurodevelopmental disorders (NDDs), including schizophrenia. Animal models have provided evidence to support these mechanistic links, with placental inflammatory responses and dysregulation of placental function implicated. This leads to changes in fetal brain cytokine balance and altered epigenetic regulation of key neurodevelopmental pathways. The prenatal timing of such mIA-evoked changes, and the accompanying fetal developmental responses to an altered in utero environment, will determine the scope of the impacts on neurodevelopmental processes. Such dysregulation can impart enduring neuropathological changes, which manifest subsequently in the postnatal period as altered neurodevelopmental behaviours in the offspring. Hence, elucidation of the functional changes that occur at the molecular level in the placenta is vital in improving our understanding of the mechanisms that underlie the pathogenesis of NDDs. This has notable relevance to the recent COVID-19 pandemic, where inflammatory responses in the placenta to SARS-CoV-2 infection during pregnancy and NDDs in early childhood have been reported. This review presents an integrated overview of these collective topics and describes the possible contribution of prenatal programming through placental effects as an underlying mechanism that links to NDD risk, underpinned by altered epigenetic regulation of neurodevelopmental pathways.
Collapse
Affiliation(s)
- Rebecca M. Woods
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jarred M. Lorusso
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jennifer Fletcher
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Heidi ElTaher
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
- Department of Physiology, Faculty of Medicine, Alexandria University, Egypt
| | - Francesca McEwan
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Isabella Harris
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Hager M. Kowash
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9WL, U.K
| | - Stephen W. D'Souza
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9WL, U.K
| | - Michael Harte
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Reinmar Hager
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jocelyn D. Glazier
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| |
Collapse
|
32
|
Delorme TC, Ozell-Landry W, Cermakian N, Srivastava LK. Behavioral and cellular responses to circadian disruption and prenatal immune activation in mice. Sci Rep 2023; 13:7791. [PMID: 37179433 PMCID: PMC10182998 DOI: 10.1038/s41598-023-34363-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
Most individuals with neurodevelopmental disorders (NDDs), including schizophrenia and autism spectrum disorders, experience disruptions in sleep and circadian rhythms. Epidemiological studies indicate that exposure to prenatal infection increases the risk of developing NDDs. We studied how environmental circadian disruption contributes to NDDs using maternal immune activation (MIA) in mice, which models prenatal infection. Pregnant dams were injected with viral mimetic poly IC (or saline) at E9.5. Adult poly IC- and saline-exposed offspring were subjected to 4 weeks of each of the following: standard lighting (LD1), constant light (LL) and standard lighting again (LD2). Behavioral tests were conducted in the last 12 days of each condition. Poly IC exposure led to significant behavioral differences, including reduced sociability (males only) and deficits in prepulse inhibition. Interestingly, poly IC exposure led to reduced sociability specifically when males were tested after LL exposure. Mice were exposed again to either LD or LL for 4 weeks and microglia were characterized. Notably, poly IC exposure led to increased microglial morphology index and density in dentate gyrus, an effect attenuated by LL exposure. Our findings highlight interactions between circadian disruption and prenatal infection, which has implications in informing the development of circadian-based therapies for individuals with NDDs.
Collapse
Affiliation(s)
- Tara C Delorme
- Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montréal, QC, H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, QC, H3A 2B4, Canada
| | - William Ozell-Landry
- Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montréal, QC, H4H 1R3, Canada
| | - Nicolas Cermakian
- Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montréal, QC, H4H 1R3, Canada.
- Department of Psychiatry, McGill University, Montréal, QC, H3A 1A1, Canada.
| | - Lalit K Srivastava
- Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montréal, QC, H4H 1R3, Canada.
- Department of Psychiatry, McGill University, Montréal, QC, H3A 1A1, Canada.
| |
Collapse
|
33
|
Hattori Y. The multifaceted roles of embryonic microglia in the developing brain. Front Cell Neurosci 2023; 17:988952. [PMID: 37252188 PMCID: PMC10213237 DOI: 10.3389/fncel.2023.988952] [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: 07/07/2022] [Accepted: 04/03/2023] [Indexed: 05/31/2023] Open
Abstract
Microglia are the resident immune cells of the central nervous system (CNS). Microglia originate from erythromyeloid progenitors in the yolk sac at the early embryonic stage, and these progenitors then colonize the CNS through extensive migration and proliferation during development. Microglia account for 10% of all cells in the adult brain, whereas the proportion of these cells in the embryonic brain is only 0.5-1.0%. Nevertheless, microglia in the developing brain widely move their cell body within the structure by extending filopodia; thus, they can interact with surrounding cells, such as neural lineage cells and vascular-structure-composing cells. This active microglial motility suggests that embryonic microglia play a pivotal role in brain development. Indeed, recent increasing evidence has revealed diverse microglial functions at the embryonic stage. For example, microglia control differentiation of neural stem cells, regulate the population size of neural progenitors and modulate the positioning and function of neurons. Moreover, microglia exert functions not only on neural lineage cells but also on blood vessels, such as supporting vascular formation and integrity. This review summarizes recent advances in the understanding of microglial cellular dynamics and multifaceted functions in the developing brain, with particular focus on the embryonic stage, and discusses the fundamental molecular mechanisms underlying their behavior.
Collapse
|
34
|
Rühle J, Ginzel M, Dietz S, Schwarz J, Lajqi T, Beer-Hammer S, Poets CF, Gille C, Köstlin-Gille N. Depletion of Ly6G-Expressing Neutrophilic Cells Leads to Altered Peripheral T-Cell Homeostasis and Thymic Development in Neonatal Mice. Int J Mol Sci 2023; 24:7763. [PMID: 37175470 PMCID: PMC10178674 DOI: 10.3390/ijms24097763] [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: 03/23/2023] [Revised: 04/14/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Newborns and especially preterm infants are much more susceptible to infections than adults. Due to immature adaptive immunity, especially innate immune cells play an important role in a newborn's infection defense. Neonatal neutrophils exhibit profound differences in their functionality compared to neutrophils of adults. In particular, neonates possess a relevant population of suppressive neutrophils, which not only inhibit but also specifically modulate the function of T-cells. In this study, we investigated whether neonatal neutrophils are already involved in T-cell development in the thymus. For this purpose, we used a newly developed model of antibody-mediated immune cell depletion in which we administered a depleting antibody to pregnant and then lactating dams. Using this method, we were able to sufficiently deplete Ly6G-positive neutrophils in offspring. We demonstrated that the depletion of neutrophils in newborn mice resulted in altered peripheral T-cell homeostasis with a decreased CD4+/CD8+ T-cell ratio and decreased expression of CD62L. Neutrophil depletion even affected T-cell development in the thymus, with increased double positive thymocytes and a decreased CD4+/CD8+ single positive thymocyte ratio. Altogether, we demonstrated a previously unknown mechanism mediating neutrophils' immunomodulatory effects in newborns.
Collapse
Affiliation(s)
- Jessica Rühle
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany; (J.R.); (S.D.); (J.S.); (C.F.P.)
| | - Marco Ginzel
- Department of Pediatric and Adolescent Surgery, Paracelsus Medical University Hospital, A-5020 Salzburg, Austria;
| | - Stefanie Dietz
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany; (J.R.); (S.D.); (J.S.); (C.F.P.)
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany; (T.L.); (C.G.)
| | - Julian Schwarz
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany; (J.R.); (S.D.); (J.S.); (C.F.P.)
| | - Trim Lajqi
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany; (T.L.); (C.G.)
| | - Sandra Beer-Hammer
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomic and ICePhA, University of Tübingen, D-72074 Tübingen, Germany;
| | - Christian F. Poets
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany; (J.R.); (S.D.); (J.S.); (C.F.P.)
| | - Christian Gille
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany; (T.L.); (C.G.)
| | - Natascha Köstlin-Gille
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany; (J.R.); (S.D.); (J.S.); (C.F.P.)
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany; (T.L.); (C.G.)
| |
Collapse
|
35
|
Gervasi MT, Romero R, Cainelli E, Veronese P, Tran MR, Jung E, Suksai M, Bosco M, Gotsch F. Intra-amniotic inflammation in the mid-trimester of pregnancy is a risk factor for neuropsychological disorders in childhood. J Perinat Med 2023; 51:363-378. [PMID: 36173676 PMCID: PMC10010737 DOI: 10.1515/jpm-2022-0255] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/17/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Intra-amniotic inflammation is a subclinical condition frequently caused by either microbial invasion of the amniotic cavity or sterile inflammatory stimuli, e.g., alarmins. An accumulating body of evidence supports a role for maternal immune activation in the genesis of fetal neuroinflammation and the occurrence of neurodevelopmental disorders such as cerebral palsy, schizophrenia, and autism. The objective of this study was to determine whether fetal exposure to mid-trimester intra-amniotic inflammation is associated with neurodevelopmental disorders in children eight to 12 years of age. METHODS This is a retrospective case-control study comprising 20 children with evidence of prenatal exposure to intra-amniotic inflammation in the mid-trimester and 20 controls matched for gestational age at amniocentesis and at delivery. Amniotic fluid samples were tested for concentrations of interleukin-6 and C-X-C motif chemokine ligand 10, for bacteria by culture and molecular microbiologic methods as well as by polymerase chain reaction for eight viruses. Neuropsychological testing of children, performed by two experienced psychologists, assessed cognitive and behavioral domains. Neuropsychological dysfunction was defined as the presence of an abnormal score (<2 standard deviations) on at least two cognitive tasks. RESULTS Neuropsychological dysfunction was present in 45% (9/20) of children exposed to intra-amniotic inflammation but in only 10% (2/20) of those in the control group (p=0.03). The relative risk (RR) of neuropsychological dysfunction conferred by amniotic fluid inflammation remained significant after adjusting for gestational age at delivery [aRR=4.5 (1.07-16.7)]. Of the 11 children diagnosed with neuropsychological dysfunction, nine were delivered at term and eight of them had mothers with intra-amniotic inflammation. Children exposed to intra-amniotic inflammation were found to have abnormalities in neuropsychological tasks evaluating complex skills, e.g., auditory attention, executive functions, and social skills, whereas the domains of reasoning, language, and memory were not affected in the cases and controls. CONCLUSIONS Asymptomatic sterile intra-amniotic inflammation in the mid-trimester of pregnancy, followed by a term birth, can still confer to the offspring a substantial risk for neurodevelopmental disorders in childhood. Early recognition and treatment of maternal immune activation in pregnancy may be a strategy for the prevention of subsequent neurodevelopmental disorders in offspring.
Collapse
Affiliation(s)
- Maria Teresa Gervasi
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA, and Detroit, MI, USA
- Gynaecology and Obstetrics Unit, Department of Women’s and Children’s Health, University Hospital of Padua, Padua, Italy
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
- Detroit Medical Center, Detroit, MI, USA
| | - Elisa Cainelli
- Department of General Psychology, University of Padova, Padova, Italy
| | - Paola Veronese
- Maternal-Fetal Medicine Unit, Department of Women’s and Children’s Health, AOPD, Padua, Italy
| | - Maria Rosa Tran
- Gynaecology and Obstetrics Unit, Department of Women’s and Children’s Health, University Hospital of Padua, Padua, Italy
| | - Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Manaphat Suksai
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Mariachiara Bosco
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Francesca Gotsch
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
36
|
Huang L, Zhan D, Xing Y, Yan Y, Li Q, Zhang J, Li S, Ning Q, Zhang C, Luo X. FGL2 deficiency alleviates maternal inflammation-induced blood-brain barrier damage by blocking PI3K/NF-κB mediated endothelial oxidative stress. Front Immunol 2023; 14:1157027. [PMID: 37051251 PMCID: PMC10083319 DOI: 10.3389/fimmu.2023.1157027] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/16/2023] [Indexed: 03/28/2023] Open
Abstract
IntroductionThe impairment of blood-brain barrier (BBB) is one of the key contributors to maternal inflammation induced brain damage in offspring. Our previous studies showed Fibrinogen-like protein 2 (FGL2) deficiency alleviated maternal inflammation induced perinatal brain damage. However, its role in BBB remains undefined.MethodsLipopolysaccharide (LPS) was intraperitoneally injected to dams at Embryonic day 17 to establish maternal inflammation model. FGL2 knockout mice and primary brain microvascular endothelial cells (BMECs) were used for the in-vivo and in-vitro experiments. BBB integrity was assessed by sodium fluorescein extravasation and tight junction (TJ) protein expression. Oxidative stress and the activation of PI3K/NF-κB pathway were evaluated to explore the mechanisms underlying.ResultsUpon maternal inflammation, BBB integrity was remarkedly reduced in neonatal mice. Meanwhile, FGL2 expression was consistently increased in BBB-impaired brain as well as in LPS-treated BMECs. Moreover, FGL2 deficiency attenuated the hyperpermeability of BBB, prevented the decline of TJ proteins, and reduced the cytokine expressions in LPS-exposed pups. Mechanistically, the indicators of oxidative stress, as well as the activation of PI3K/NF-κB pathway, were upregulated after LPS exposure in vivo and in vitro. FGL2 deletion decreased the generation of ROS and NO, reduced the endothelial iNOS and NOX2 expressions, and suppressed the PI3K/NF-κB pathway activation. Besides, inhibition of PI3K by LY294002 decreased the oxidative stress in LPS-treated wild-type BMECs. While, overexpression of PI3K by lentivirus reemerged the induction of NOX2 and iNOS as well as NF-κB activation in FGL2-deleted BMECs.ConclusionOur findings indicate that FGL2 deficiency alleviates the maternal inflammation-induced BBB disruption by inhibiting PI3K/NF-κB mediated oxidative stress in BMECs. Targeting FGL2 may provide a new therapy for prenatal brain damage of offspring.
Collapse
Affiliation(s)
- Lianjing Huang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Di Zhan
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ying Xing
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yaqin Yan
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qing Li
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jingyi Zhang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Sujuan Li
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qin Ning
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Cai Zhang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- *Correspondence: Xiaoping Luo, ; Cai Zhang,
| | - Xiaoping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- *Correspondence: Xiaoping Luo, ; Cai Zhang,
| |
Collapse
|
37
|
Jaber M. Genetic and environmental mouse models of autism reproduce the spectrum of the disease. J Neural Transm (Vienna) 2023; 130:425-432. [PMID: 36318343 DOI: 10.1007/s00702-022-02555-9] [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: 06/28/2022] [Accepted: 10/17/2022] [Indexed: 03/23/2023]
Abstract
Genetic and environmental factors increase autism spectrum disorder (ASD) incidence, and this has led to the generation of corresponding animal models, with some showing strong construct and face validity. This short review focuses on results we have recently obtained with environmental and genetic mouse models of ASD and that are the valproic acid, the poly I:C and the Shank 3 models. This has allowed us to provide a comparative description of these widely used animal models providing an interesting perspective as to the pros and cons of each one of them, in our experimental settings. In these papers, we focused on motor and gait disorders which are currently not included in the diagnosis criteria, but which may provide new insights to ASD pathophysiology potentially leading to innovative therapies for a disease that currently has none. In all these models, we reported behavioral, cellular and molecular alterations related to the cerebellum. Motor and gait deficits were observed to various degrees in animal models and, when strongly present, they were correlated to the severity of social deficits as well as to the number of cerebellar Purkinje cells. Additionally, we also reported that, like in humans, males are more severely affected than females in these ASD models. These findings, along with an increasing body of literature, open new hopes in the ASD field pointing to brain regions, such the cerebellum, that are at the crossroads between cognitive, social and motor deficits. Targeting these brain regions and their underlying pathways and synaptic connections may prove of significant benefits.
Collapse
Affiliation(s)
- Mohamed Jaber
- Université de Poitiers, Inserm, Laboratoire de Neurosciences Expérimentales et Cliniques, Bâtiment B36, 1 Rue Georges Bonnet, BP 633, TSA 51106, 86073, Poitiers cedex9, France.
- Centre Hospitalier Universitaire de Poitiers, Poitiers, France.
| |
Collapse
|
38
|
Microbiota-immune-brain interactions: A lifespan perspective. Curr Opin Neurobiol 2023; 78:102652. [PMID: 36463579 DOI: 10.1016/j.conb.2022.102652] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/10/2022] [Accepted: 10/31/2022] [Indexed: 12/03/2022]
Abstract
There is growing appreciation of key roles of the gut microbiota in maintaining homeostasis and influencing brain and behaviour at critical windows across the lifespan. Mounting evidence suggests that communication between the gut and the brain could be the key to understanding multiple neuropsychiatric disorders, with the immune system coming to the forefront as an important mechanistic mediator. Throughout the lifespan, the immune system exchanges continuous reciprocal signals with the central nervous system. Intestinal microbial cues alter immune mediators with consequences for host neurophysiology and behaviour. Several factors challenge the gut microbiota composition, which in response release molecules with neuro- and immuno-active potential that are crucial for adequate neuro-immune interactions. In this review, multiple factors contributing to the upkeep of the fine balance between health and disease of these systems are discussed, and we elucidate the potential mechanistic implications for the gut microbiota inputs on host brain and behaviour across the lifespan.
Collapse
|
39
|
Relationship between the Dietary Inflammatory Index Score and Cytokine Levels in Chinese Pregnant Women during the Second and Third Trimesters. Nutrients 2022; 15:nu15010194. [PMID: 36615851 PMCID: PMC9824482 DOI: 10.3390/nu15010194] [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: 10/27/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
The impact of dietary inflammatory potential on serum cytokine concentrations in second and third trimesters of Chinese pregnant women is not clear. A total of 175 pregnant women from the Tianjin Maternal and Child Health Education and Service Cohort (TMCHESC) were included. The dietary inflammatory index (DII) was calculated based on 24-h food records. Serum tumor necrosis factor-α (TNF-α), interleukin 1β (IL-1β), IL-6, IL-8, IL-10, C-reactive protein (CRP), and monocyte chemoattractant protein-1 (MCP-1) levels in the second and third trimesters were measured. The mean DII scores (mean ± SD) were -0.07 ± 1.65 and 0.06 ± 1.65 in the second and third trimesters, respectively. In the third trimester, IL-1β (p = 0.039) and MCP-1 (p = 0.035) levels decreased and then increased with increasing DII scores. IL-10 concentrations decreased in pregnant women whose DII scores increased between the second and third trimesters (p = 0.011). Thiamin and vitamin C were negatively correlated with MCP-1 (β = -0.879, and β = -0.003) and IL-6 (β = -0.602, and β = -0.002) levels in the third trimester. In conclusion, the DII score had a U-shaped association with cytokine levels during the third trimester. Changes in DII scores between the second and third trimesters of pregnancy were correlated with cytokine levels during the third trimester.
Collapse
|
40
|
Lee K, Mills Z, Cheung P, Cheyne JE, Montgomery JM. The Role of Zinc and NMDA Receptors in Autism Spectrum Disorders. Pharmaceuticals (Basel) 2022; 16:ph16010001. [PMID: 36678498 PMCID: PMC9866730 DOI: 10.3390/ph16010001] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
NMDA-type glutamate receptors are critical for synaptic plasticity in the central nervous system. Their unique properties and age-dependent arrangement of subunit types underpin their role as a coincidence detector of pre- and postsynaptic activity during brain development and maturation. NMDAR function is highly modulated by zinc, which is co-released with glutamate and concentrates in postsynaptic spines. Both NMDARs and zinc have been strongly linked to autism spectrum disorders (ASDs), suggesting that NMDARs are an important player in the beneficial effects observed with zinc in both animal models and children with ASDs. Significant evidence is emerging that these beneficial effects occur via zinc-dependent regulation of SHANK proteins, which form the backbone of the postsynaptic density. For example, dietary zinc supplementation enhances SHANK2 or SHANK3 synaptic recruitment and rescues NMDAR deficits and hypofunction in Shank3ex13-16-/- and Tbr1+/- ASD mice. Across multiple studies, synaptic changes occur in parallel with a reversal of ASD-associated behaviours, highlighting the zinc-dependent regulation of NMDARs and glutamatergic synapses as therapeutic targets for severe forms of ASDs, either pre- or postnatally. The data from rodent models set a strong foundation for future translational studies in human cells and people affected by ASDs.
Collapse
|
41
|
Zhang X. Magnetic resonance imaging of the monkey fetal brain in utero. INVESTIGATIVE MAGNETIC RESONANCE IMAGING 2022; 26:177-190. [PMID: 36937817 PMCID: PMC10019598 DOI: 10.13104/imri.2022.26.4.177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Non-human primates (NHPs) are the closest living relatives of the human and play a critical role in investigating the effects of maternal viral infection and consumption of medicines, drugs, and alcohol on fetal development. With the advance of contemporary fast MRI techniques with parallel imaging, fetal MRI is becoming a robust tool increasingly used in clinical practice and preclinical studies to examine congenital abnormalities including placental dysfunction, congenital heart disease (CHD), and brain abnormalities non-invasively. Because NHPs are usually scanned under anesthesia, the motion artifact is reduced substantially, allowing multi-parameter MRI techniques to be used intensively to examine the fetal development in a single scanning session or longitudinal studies. In this paper, the MRI techniques for scanning monkey fetal brains in utero in biomedical research are summarized. Also, a fast imaging protocol including T2-weighted imaging, diffusion MRI, resting-state functional MRI (rsfMRI) to examine rhesus monkey fetal brains in utero on a clinical 3T scanner is introduced.
Collapse
Affiliation(s)
- Xiaodong Zhang
- EPC Imaging Center and Division of Neuropharmacology and Neurologic Diseases, Emory National Primate Research Center, Emory University, Atlanta, Georgia, 30329, USA
| |
Collapse
|
42
|
Cipriani C, Tartaglione AM, Giudice M, D’Avorio E, Petrone V, Toschi N, Chiarotti F, Miele MT, Calamandrei G, Garaci E, Matteucci C, Sinibaldi-Vallebona P, Ricceri L, Balestrieri E. Differential Expression of Endogenous Retroviruses and Inflammatory Mediators in Female and Male Offspring in a Mouse Model of Maternal Immune Activation. Int J Mol Sci 2022; 23:13930. [PMID: 36430402 PMCID: PMC9695919 DOI: 10.3390/ijms232213930] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/16/2022] Open
Abstract
Maternal infections during pregnancy and the consequent maternal immune activation (MIA) are the major risk factors for autism spectrum disorder (ASD). Epidemiological evidence is corroborated by the preclinical models in which MIA leads to ASD-like behavioral abnormalities and altered neuroinflammatory profiles, with an increase in pro-inflammatory cytokines and microglial markers. In addition to neuroinflammatory response, an abnormal expression of endogenous retroviruses (ERVs) has been identified in neurodevelopmental disorders and have been found to correlate with disease severity. Our aim was to evaluate the transcriptional profile of several ERV families, ERV-related genes, and inflammatory mediators (by RT real-time PCR) in mouse offspring of both sexes, prenatally exposed to polyinosinic:polycytidylic acid (Poly I:C), a synthetic double-stranded RNA molecule targeting TLR-3 that mimics viral maternal infection during pregnancy. We found that prenatal exposure to Poly I:C deregulated the expression of some ERVs and ERV-related genes both in the prefrontal cortex (PFC) and hippocampus, while no changes were detected in the blood. Interestingly, sex-related differences in the expression levels of some ERVs, ERV-related genes, and inflammatory mediators that were higher in females than in males emerged only in PFC. Our findings support the tissue specificity of ERV and ERV-related transcriptional profiles in MIA mice.
Collapse
Affiliation(s)
- Chiara Cipriani
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Anna Maria Tartaglione
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità (ISS), 00161 Rome, Italy
| | - Martina Giudice
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Erica D’Avorio
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Vita Petrone
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Nicola Toschi
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy
- Martinos Center for Biomedical Imaging, Harvard Medical School, Boston, MA 02115, USA
| | - Flavia Chiarotti
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità (ISS), 00161 Rome, Italy
| | - Martino Tony Miele
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Gemma Calamandrei
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità (ISS), 00161 Rome, Italy
| | - Enrico Garaci
- University San Raffaele, 00166 Rome, Italy
- IRCCS San Raffaele Pisana, 00163 Rome, Italy
| | - Claudia Matteucci
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Paola Sinibaldi-Vallebona
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
- Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy
| | - Laura Ricceri
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità (ISS), 00161 Rome, Italy
| | - Emanuela Balestrieri
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| |
Collapse
|
43
|
Griego E, Segura-Villalobos D, Lamas M, Galván EJ. Maternal immune activation increases excitability via downregulation of A-type potassium channels and reduces dendritic complexity of hippocampal neurons of the offspring. Brain Behav Immun 2022; 105:67-81. [PMID: 35803480 DOI: 10.1016/j.bbi.2022.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/29/2022] [Accepted: 07/03/2022] [Indexed: 11/29/2022] Open
Abstract
The epidemiological association between bacterial or viral maternal infections during pregnancy and increased risk for developing psychiatric disorders in offspring is well documented. Numerous rodent and non-human primate studies of viral- or, to a lesser extent, bacterial-induced maternal immune activation (MIA) have documented a series of neurological alterations that may contribute to understanding the pathophysiology of schizophrenia and autism spectrum disorders. Long-term neuronal and behavioral alterations are now ascribed to the effect of maternal proinflammatory cytokines rather than the infection itself. However, detailed electrophysiological alterations in brain areas relevant to psychiatric disorders, such as the dorsal hippocampus, are lacking in response to bacterial-induced MIA. This study determined if electrophysiological and morphological alterations converge in CA1 pyramidal cells (CA1 PC) from the dorsal hippocampus in bacterial-induced MIA offspring. A series of changes in the functional expression of K+ and Na+ ion channels altered the passive and active membrane properties and triggered hyperexcitability of CA1 PC. Contributing to the hyperexcitability, the somatic A-type potassium current (IA) was decreased in MIA CA1 PC. Likewise, the spontaneous glutamatergic and GABAergic inputs were dysregulated and biased toward increased excitation, thereby reshaping the excitation-inhibition balance. Consistent with these findings, the dendritic branching complexity of MIA CA1 PC was reduced. Together, these morphophysiological alterations modify CA1 PC computational capabilities and contribute to explaining cellular alterations that may underlie the cognitive symptoms of MIA-associated psychiatric disorders.
Collapse
Affiliation(s)
- Ernesto Griego
- Departamento de Farmacobiología, CINVESTAV Unidad Sur, Ciudad de México, Mexico
| | | | - Mónica Lamas
- Departamento de Farmacobiología, CINVESTAV Unidad Sur, Ciudad de México, Mexico
| | - Emilio J Galván
- Departamento de Farmacobiología, CINVESTAV Unidad Sur, Ciudad de México, Mexico.
| |
Collapse
|
44
|
Otero AM, Antonson AM. At the crux of maternal immune activation: Viruses, microglia, microbes, and IL-17A. Immunol Rev 2022; 311:205-223. [PMID: 35979731 PMCID: PMC9804202 DOI: 10.1111/imr.13125] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Inflammation during prenatal development can be detrimental to neurodevelopmental processes, increasing the risk of neuropsychiatric disorders. Prenatal exposure to maternal viral infection during pregnancy is a leading environmental risk factor for manifestation of these disorders. Preclinical animal models of maternal immune activation (MIA), established to investigate this link, have revealed common immune and microbial signaling pathways that link mother and fetus and set the tone for prenatal neurodevelopment. In particular, maternal intestinal T helper 17 cells, educated by endogenous microbes, appear to be key drivers of effector IL-17A signals capable of reaching the fetal brain and causing neuropathologies. Fetal microglial cells are particularly sensitive to maternally derived inflammatory and microbial signals, and they shift their functional phenotype in response to MIA. Resulting cortical malformations and miswired interneuron circuits cause aberrant offspring behaviors that recapitulate core symptoms of human neurodevelopmental disorders. Still, the popular use of "sterile" immunostimulants to initiate MIA has limited translation to the clinic, as these stimulants fail to capture biologically relevant innate and adaptive inflammatory sequelae induced by live pathogen infection. Thus, there is a need for more translatable MIA models, with a focus on relevant pathogens like seasonal influenza viruses.
Collapse
Affiliation(s)
- Ashley M. Otero
- Neuroscience ProgramUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Adrienne M. Antonson
- Department of Animal SciencesUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| |
Collapse
|
45
|
Park HJ, Jung H. Neuro-immune interactions at single-cell resolution in neurodevelopmental, infectious, and neurodegenerative diseases. Anim Cells Syst (Seoul) 2022; 26:137-147. [PMID: 36046030 PMCID: PMC9423835 DOI: 10.1080/19768354.2022.2110937] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Recent technological advance in single-cell and single-nucleus transcriptomics has made it possible to generate an unprecedentedly detailed landscape of neuro-immune interactions in healthy and diseased brains. In this review, we overview the recent literature that catalogs single-cell-level gene expression in brains with signs of inflammation, focusing on maternal immune activation, viral infection, and auto-immune diseases. The literature also includes a series of papers that provide strong evidence for immunological contributions to neurodegenerative diseases, which, in a strict sense, are not considered neuroinflammatory. To help with the discussion, we present a diagram of experimental and analytical flows in the single-cell analysis of the brain. We also discuss the recurring themes of neuro-immune interactions and suggest future research directions.
Collapse
Affiliation(s)
- Hyun Jung Park
- Samsung Medical Center, Samsung Genome Institute, Seoul, Republic of Korea
| | - Hosung Jung
- Department of Anatomy, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
46
|
Jensen AR, Lane AL, Werner BA, McLees SE, Fletcher TS, Frye RE. Modern Biomarkers for Autism Spectrum Disorder: Future Directions. Mol Diagn Ther 2022; 26:483-495. [PMID: 35759118 PMCID: PMC9411091 DOI: 10.1007/s40291-022-00600-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2022] [Indexed: 11/19/2022]
Abstract
Autism spectrum disorder is an increasingly prevalent neurodevelopmental disorder in the world today, with an estimated 2% of the population being affected in the USA. A major complicating factor in diagnosing, treating, and understanding autism spectrum disorder is that defining the disorder is solely based on the observation of behavior. Thus, recent research has focused on identifying specific biological abnormalities in autism spectrum disorder that can provide clues to diagnosis and treatment. Biomarkers are an objective way to identify and measure biological abnormalities for diagnostic purposes as well as to measure changes resulting from treatment. This current opinion paper discusses the state of research of various biomarkers currently in development for autism spectrum disorder. The types of biomarkers identified include prenatal history, genetics, neurological including neuroimaging, neurophysiologic, and visual attention, metabolic including abnormalities in mitochondrial, folate, trans-methylation, and trans-sulfuration pathways, immune including autoantibodies and cytokine dysregulation, autonomic nervous system, and nutritional. Many of these biomarkers have promising preliminary evidence for prenatal and post-natal pre-symptomatic risk assessment, confirmation of diagnosis, subtyping, and treatment response. However, most biomarkers have not undergone validation studies and most studies do not investigate biomarkers with clinically relevant comparison groups. Although the field of biomarker research in autism spectrum disorder is promising, it appears that it is currently in the early stages of development.
Collapse
Affiliation(s)
- Amanda R Jensen
- Section on Neurodevelopmental Disorders, Barrow Neurological Institute at Phoenix Children's Hospital, 1919 E Thomas Rd, Phoenix, AZ, 85016, USA
| | - Alison L Lane
- Section on Neurodevelopmental Disorders, Barrow Neurological Institute at Phoenix Children's Hospital, 1919 E Thomas Rd, Phoenix, AZ, 85016, USA
| | - Brianna A Werner
- Section on Neurodevelopmental Disorders, Barrow Neurological Institute at Phoenix Children's Hospital, 1919 E Thomas Rd, Phoenix, AZ, 85016, USA
| | - Sallie E McLees
- Section on Neurodevelopmental Disorders, Barrow Neurological Institute at Phoenix Children's Hospital, 1919 E Thomas Rd, Phoenix, AZ, 85016, USA
| | - Tessa S Fletcher
- Section on Neurodevelopmental Disorders, Barrow Neurological Institute at Phoenix Children's Hospital, 1919 E Thomas Rd, Phoenix, AZ, 85016, USA.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Richard E Frye
- Section on Neurodevelopmental Disorders, Barrow Neurological Institute at Phoenix Children's Hospital, 1919 E Thomas Rd, Phoenix, AZ, 85016, USA.
| |
Collapse
|