1
|
Shimamura T, Kitashiba M, Nishizawa K, Hattori Y. Physiological roles of embryonic microglia and their perturbation by maternal inflammation. Front Cell Neurosci 2025; 19:1552241. [PMID: 40260079 PMCID: PMC12009865 DOI: 10.3389/fncel.2025.1552241] [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: 12/27/2024] [Accepted: 03/24/2025] [Indexed: 04/23/2025] Open
Abstract
The interplay between the nervous and immune systems is well documented in the context of adult physiology and disease. Recent advances in understanding immune cell development have highlighted a significant interaction between neural lineage cells and microglia, the resident brain macrophages, during developmental stages. Throughout development, particularly from the embryonic to postnatal stages, diverse neural lineage cells are sequentially generated, undergo fate determination, migrate dynamically to their appropriate locations while maturing, and establish connections with their surroundings to form neural circuits. Previous studies have demonstrated that microglia contribute to this highly orchestrated process, ensuring the proper organization of brain structure. These findings underscore the need to further investigate how microglia behave and function within a broader framework of neurodevelopment. Importantly, recent epidemiological studies have suggested that maternal immune activation (MIA), triggered by various factors, such as viral or bacterial infections, environmental stressors, or other external influences, can affect neurogenesis and neural circuit formation, increasing the risk of neurodevelopmental disorders (NDDs) in offspring. Notably, many studies have revealed that fetal microglia undergo significant changes in response to MIA. Given their essential roles in neurogenesis and vascular development, inappropriate activation or disruption of microglial function may impair these critical processes, potentially leading to abnormal neurodevelopment. This review highlights recent advances in rodent models and human studies that have shed light on the behaviors and multifaceted roles of microglia during brain development, with a particular focus on the embryonic stage. Furthermore, drawing on insights from rodent MIA models, this review explores how MIA disrupts microglial function and how such disturbances may impair brain development, ultimately contributing to the onset of NDDs.
Collapse
Affiliation(s)
| | | | | | - Yuki Hattori
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| |
Collapse
|
2
|
Viana-de-Lima L, Platt N, Zamaro IHO, Karasiak GD, Kaster MP. A Comprehensive Review of poly(I: C) as a Tool for Investigating Astrocytic TLR3 Signaling. Neurochem Res 2025; 50:133. [PMID: 40172723 DOI: 10.1007/s11064-025-04381-3] [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: 01/23/2025] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 04/04/2025]
Abstract
Astrocytes play a crucial role in regulating the structure, function, and interactions between the synaptic and vascular compartments in the brain. Toll-like receptor 3 (TLR3) is expressed in astrocytes and recognizes double-stranded RNA (dsRNA), a pathogen-associated molecular pattern (PAMP). This review examines the current understanding of TLR3 signaling, with a focus on its specific role in astrocytes, and the use of the viral mimetic polyinosinic: polycytidylic acid (poly(I: C)) to model the effects of viral infections in both in vitro and in vivo studies. Poly(I: C) is a useful tool for studying neuro-immune communication and has significantly added to our knowledge of how the brain responds to immune challenges. Upon poly(I: C) exposure, TLR3 activation in astrocytes triggers inflammatory signaling pathways, leading to both antiviral responses and neuroinflammation. However, further research is required to investigate the cell-specific impacts of TLR3 activation, along with the influence of developmental stages, brain regions, and sex-specific responses, to gain a comprehensive understanding of how immune activation shapes the development and function of the central nervous system (CNS).
Collapse
Affiliation(s)
- Leonardo Viana-de-Lima
- Laboratory of Translational Neuroscience, Department of Biochemistry, Federal University of Santa Catarina (UFSC), Florianopolis, Santa Catarina, Brazil
| | - Nicolle Platt
- Laboratory of Translational Neuroscience, Department of Biochemistry, Federal University of Santa Catarina (UFSC), Florianopolis, Santa Catarina, Brazil
| | - Isabele Haruna Ono Zamaro
- Laboratory of Translational Neuroscience, Department of Biochemistry, Federal University of Santa Catarina (UFSC), Florianopolis, Santa Catarina, Brazil
| | - Gabriela Duarte Karasiak
- Laboratory of Translational Neuroscience, Department of Biochemistry, Federal University of Santa Catarina (UFSC), Florianopolis, Santa Catarina, Brazil
| | - Manuella Pinto Kaster
- Laboratory of Translational Neuroscience, Department of Biochemistry, Federal University of Santa Catarina (UFSC), Florianopolis, Santa Catarina, Brazil.
| |
Collapse
|
3
|
Biggar E, Thomas R, Lave ML, Jaju Bhattad G, Rajakumar N, Renaud SJ. Maternal immune activation elicits rapid and sex-dependent changes in gene expression and vascular dysfunction in the rat placenta. Placenta 2025; 163:51-60. [PMID: 40081234 DOI: 10.1016/j.placenta.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 02/12/2025] [Accepted: 03/01/2025] [Indexed: 03/15/2025]
Abstract
INTRODUCTION Maternal immune activation (MIA), characterized by increased circulating inflammatory mediators during pregnancy, is associated with adverse pregnancy outcomes and neurodevelopmental deficits in offspring. These health outcomes often manifest differently depending on fetal-placental sex. A well-established model of MIA involves administration of a viral mimetic, polyinosinic:polycytidilic acid (PolyI:C), to pregnant rodents. Placental responses to PolyI:C contribute to the detrimental effects of MIA on offspring, but these responses have not yet been well characterized. In the present study, we profiled acute gene expression changes in male and female placentas following PolyI:C administration to pregnant rats during late gestation. METHODS Pregnant rats received 4 mg/kg PolyI:C or saline intravenously on gestational day 18.5, and tissues were harvested 4-5 h later. Gene expression profiling on placental tissue was performed. Enzyme immunoassays and immunohistochemistry were conducted to determine levels of select proteins in maternal blood and placental tissue, respectively. RESULTS Maternal PolyI:C exposure caused a robust increase in levels of inflammatory mediators in maternal blood and placental tissue. There were more genes differentially expressed in female placentas after PolyI:C exposure (765) than male placentas (221), including reduced expression of genes associated with maternal-fetal communication. Placentas also had increased expression of genes linked with vascular dysfunction after PolyI:C-induced MIA. DISCUSSION PolyI:C elicited a powerful inflammatory response in the placenta along with vascular dysfunction, likely contributing to the adverse pregnancy outcomes triggered by MIA. Female placentas responded to PolyI:C more vigorously than male placentas, which could underlie the differential outcomes of MIA depending on sex.
Collapse
Affiliation(s)
- Erin Biggar
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, MSB428, 1151 Richmond Street, London, ON, N6A 5C1, Canada
| | - Ruth Thomas
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, MSB428, 1151 Richmond Street, London, ON, N6A 5C1, Canada
| | - Megan L Lave
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, MSB428, 1151 Richmond Street, London, ON, N6A 5C1, Canada
| | - Gargi Jaju Bhattad
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, MSB428, 1151 Richmond Street, London, ON, N6A 5C1, Canada
| | - Nagalingam Rajakumar
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, MSB428, 1151 Richmond Street, London, ON, N6A 5C1, Canada
| | - Stephen J Renaud
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, MSB428, 1151 Richmond Street, London, ON, N6A 5C1, Canada; Children's Health Research Institute, London Health Sciences Centre Research Institute, London, ON, Canada.
| |
Collapse
|
4
|
Delorme TC, Arcego DM, Penichet D, O'Toole N, Huebener N, Silveira PP, Srivastava LK, Cermakian N. Large-scale effects of prenatal inflammation and early life circadian disruption in mice: Implications for neurodevelopmental disorders. Brain Behav Immun 2025; 127:409-422. [PMID: 40118225 DOI: 10.1016/j.bbi.2025.03.023] [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: 11/17/2024] [Revised: 02/17/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025] Open
Abstract
Around 80 % of individuals with neurodevelopmental disorders such as schizophrenia and autism spectrum disorders experience disruptions in sleep/circadian rhythms. We explored whether environmental circadian disruption interacts with prenatal infection, a risk factor for neurodevelopmental disorders, to induce sex-specific deficits in mice. A maternal immune activation (MIA) protocol was used by injecting pregnant mice with viral mimic poly IC or saline at E9.5. Juvenile/adolescent male and female offspring (3-7 weeks old) were then subjected to a standard light:dark cycle (12:12LD) or to constant light (LL). Significant interactions between treatment (MIA, control) and lighting (12:12LD, LL) were evident in behaviors related to cognition, anxiety, and sociability. This pattern persisted in our RNA sequencing analysis of the dorsal hippocampus, where poly IC exposure resulted in numerous differentially expressed genes (DEGs) in males, while exposure to both poly IC and LL led to a marked reduction in DEGs. Through WGCNA analysis, many significant gene modules were found to be positively associated with poly IC (vs. saline) and LL (vs. LD) in males (fewer in females). Many of the identified hub-bottleneck genes were homologous to human genes associated with sleep/circadian rhythms and neurodevelopmental disorders as revealed by GWA studies. The MIA- and LL-associated modules were enriched in microglia gene signatures, which was paralleled by trends of effects of each of the factors on microglia morphology. In conclusion, in a mouse model of prenatal infection, circadian disruption induced by LL during adolescence acts as a modulator of the effects of MIA at behavioral, cellular, and molecular levels.
Collapse
Affiliation(s)
- Tara C Delorme
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Integrated Program in Neuroscience, McGill University, Montréal, Québec H3A 2B4, Canada
| | - Danusa M Arcego
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Department of Psychiatry, McGill University, Montréal, Québec H3A 1A1, Canada
| | - Danae Penichet
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Integrated Program in Neuroscience, McGill University, Montréal, Québec H3A 2B4, Canada
| | - Nicholas O'Toole
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada
| | - Nikki Huebener
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada
| | - Patrícia P Silveira
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Department of Psychiatry, McGill University, Montréal, Québec H3A 1A1, Canada
| | - Lalit K Srivastava
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Department of Psychiatry, McGill University, Montréal, Québec H3A 1A1, Canada.
| | - Nicolas Cermakian
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Department of Psychiatry, McGill University, Montréal, Québec H3A 1A1, Canada.
| |
Collapse
|
5
|
Scott KJ, Bilkey DK. Sex-dependent effects of rat maternal immune activation on motor function in offspring of poly I:C treated rats. Behav Brain Res 2025; 481:115431. [PMID: 39814236 DOI: 10.1016/j.bbr.2025.115431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 01/18/2025]
Abstract
A majority of people with schizophrenia will experience motor symptoms such as impairments to coordination, balance and motor sequencing. These neurological soft signs are associated with negative social and functional outcomes, and poor disease prognosis. They occur prior to medication exposure, suggesting they are an intrinsic feature of schizophrenia. Despite the need to better understand this dysfunction, relatively few studies have provided a detailed focus on motor capability in animal models of schizophrenia. Here we investigate motor coordination in a rat maternal immune activation (MIA) model of schizophrenia risk. The female and male offspring of Polyinosinic:polycytidylic acid (Poly I:C), and vehicle-treated, pregnant dams were tested in a horizontal ladder rung task using regular and irregular rung configurations. We extracted information about limb positions from video, and measured faults and gait coordination in the task. We found that adult male MIA rats were more likely to slip from the ladder rungs than control animals, and they were more likely to have multiple limbs slip simultaneously. MIA rats also exhibited more variability in stride length, a result that correlated with slips and mirrored disease-related changes in human gait. In contrast, female MIA rats displayed minimal alterations in motor performance. Our findings show that the ladder task uncovers sex-dependent effects on motor coordination in MIA rats and highlights the potential usefulness of the MIA model for investigating motor dysfunction in an animal model of schizophrenia risk.
Collapse
Affiliation(s)
- K Jack Scott
- Department of Psychology, University of Otago, New Zealand
| | - David K Bilkey
- Department of Psychology, University of Otago, New Zealand.
| |
Collapse
|
6
|
Davis LK, Ince LM, Gullapalli S, Fonken LK. Neuroimmune and behavioral changes elicited by maternal immune activation in mice are ameliorated by early postnatal immune stimulation. Brain Behav Immun 2025; 127:375-386. [PMID: 40081778 DOI: 10.1016/j.bbi.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025] Open
Abstract
Though the etiology of autism spectrum disorder (ASD) is complex and not fully understood, it is believed that genetic risk factors, coupled with early life inflammation may predispose individuals to develop ASD. Maternal immune activation (MIA) is associated with increased incidence of ASD in offspring; however, not all mothers who experience inflammation during pregnancy have children with autism, suggesting that MIA may act as a disease primer that results in ASD pathology when paired with additional inflammatory insults. Here, we tested the hypothesis that MIA is a disease primer by using a two-hit model that combined MIA with a secondary immune stimulation in early life. C57BL/6J mouse dams were treated with polyinosinic-polycytidylic acid (Poly(I:C)) at embyronic day 12.5, and a subset of litters were then treated with the endotoxin lipopolysaccharide (LPS) four days after birth. Offspring were assessed in young adulthood for changes in behavior including sociability, repetitive-like behaviors, and anxiety-like behaviors. Flow cytometry was performed in adulthood to assess changes in immune cell populations in the periphery and in the brain. MIA increased repetitive-like behaviors in male mice and decreased sociability in both sexes. Unexpectedly, the secondary immune stimulation with LPS did not exacerbate changes in social and repetitive-like behaviors in either sex. MIA also altered distribution of cytotoxic CD8 + T cell populations in the periphery and brain of both sexes: CD8 + T cells were elevated in thymus but reduced in spleen, lymph, and brain. Additionally, MIA altered microglia activity in a region-specific manner in male mice, which was also not exacerbated but rather ameliorated when combined with LPS. Our results demonstrate that changes in repetitive-like and social behaviors that are induced by MIA in male mice are not exacerbated by subsequent inflammatory challenge and highlights the importance of considering the timing of stressors in the appearance of developmental pathology.
Collapse
Affiliation(s)
- Lourdes K Davis
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA; Interdisciplinary Neuroscience Program, University of Texas at Austin, Austin, TX 78712, USA.
| | - Louise M Ince
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Sriya Gullapalli
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Laura K Fonken
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA; Interdisciplinary Neuroscience Program, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
7
|
Hall MB, Lemanski EA, Schwarz JM. Prenatal Maternal Immune Activation with Lipopolysaccharide Accelerates the Developmental Acquisition of Neonatal Reflexes in Rat Offspring Without Affecting Maternal Care Behaviors. Biomolecules 2025; 15:347. [PMID: 40149883 PMCID: PMC11940702 DOI: 10.3390/biom15030347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 02/18/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025] Open
Abstract
Maternal immune activation (MIA)-infection with an immunogen during pregnancy-is linked to an increased risk of neurodevelopmental disorders (NDDs) in offspring. Both MIA and NDDs are associated with developmental delays in offsprings' motor behavior. Therefore, the current study examined the effects of MIA on neonatal reflex development in male and female offspring. Sprague Dawley rats were administered lipopolysaccharide (LPS; 50 μg/mL/kg, i.p.) or saline on embryonic day (E)15 of gestation. The offspring were then tested daily from postnatal day (P)3-P21 to determine their neonatal reflex abilities. The maternal care behaviors of the dam were also quantified on P1-P5, P10, and P15. We found that, regardless of sex, the E15 LPS offspring were able to forelimb grasp, cliff avoid, and right with a correct posture at an earlier postnatal age than the E15 saline offspring did. The E15 LPS offspring also showed better performance of forelimb grasping, hindlimb grasping, righting with correct posture, and walking with correct posture than the E15 saline offspring did. There were no significant differences in maternal licking/grooming, arched-back nursing, non-arched-back nursing, or total nursing across the E15 groups. Overall, these findings suggest that MIA with LPS on E15 accelerates reflex development in offspring without affecting maternal care. This may be explained by the stress acceleration hypothesis, whereby early-life stress accelerates development to promote survival.
Collapse
Affiliation(s)
- Mary Beth Hall
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA;
- Interdisciplinary Neuroscience Graduate Program, University of Delaware, Newark, DE 19716, USA
| | - Elise A. Lemanski
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA;
- Interdisciplinary Neuroscience Graduate Program, University of Delaware, Newark, DE 19716, USA
| | - Jaclyn M. Schwarz
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA;
- Interdisciplinary Neuroscience Graduate Program, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
8
|
Martín-Guerrero SM, Martín-Estebané M, Lara Ordóñez AJ, Cánovas M, Martín-Oliva D, González-Maeso J, Cutillas PR, López-Giménez JF. Maternal immune activation imprints translational dysregulation and differential MAP2 phosphorylation in descendant neural stem cells. Mol Psychiatry 2025:10.1038/s41380-025-02905-5. [PMID: 39900676 DOI: 10.1038/s41380-025-02905-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 12/19/2024] [Accepted: 01/21/2025] [Indexed: 02/05/2025]
Abstract
Alterations induced by maternal immune activation (MIA) during gestation impact the subsequent neurodevelopment of progeny, a process that in humans, has been linked to the development of several neuropsychiatric conditions. To undertake a comprehensive examination of the molecular mechanisms governing MIA, we have devised an in vitro model based on neural stem cells (NSCs) sourced from fetuses carried by animals subjected to Poly I:C treatment. These neural progenitors demonstrate proliferative capacity and can be effectively differentiated into both neurons and glial cells. Transcriptomic, proteomic, and phosphoproteomic analyses conducted on these cellular models, in conjunction with counterparts from control treatments, revealed discernible shifts in the expression levels of a specific subset of proteins implicated in neuronal function. Furthermore, the phosphoproteomic data highlighted a discernible discrepancy in the basal phosphorylation of proteins between differentiated cells from both experimental groups, particularly within proteins associated with cytoskeletal architecture and synaptic functionality, notably those belonging to the MAP family. Observed alterations in MAP phosphorylation were found to potentially have functional consequences as they correlate with changes in neuronal plasticity and the establishment of neuronal synapses. Our data agrees with previous published observations and further underscore the importance of MAP2 phosphorylation state on its function and the impact that this protein has in neuronal structure and function.
Collapse
Affiliation(s)
- Sandra M Martín-Guerrero
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK
| | | | | | - Miguel Cánovas
- Instituto de Parasitología y Biomedicina "López-Neyra" (IPBLN-CSIC), Granada, Spain
| | - David Martín-Oliva
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - Javier González-Maeso
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Pedro R Cutillas
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK.
| | - Juan F López-Giménez
- Instituto de Parasitología y Biomedicina "López-Neyra" (IPBLN-CSIC), Granada, Spain.
| |
Collapse
|
9
|
Chang CY, Dai W, Hu SSJ. Cannabidiol enhances socially transmitted food preference: a role of acetylcholine in the mouse basal forebrain. Psychopharmacology (Berl) 2025; 242:247-269. [PMID: 39158618 DOI: 10.1007/s00213-024-06670-1] [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: 05/19/2024] [Accepted: 08/05/2024] [Indexed: 08/20/2024]
Abstract
RATIONALE AND OBJECTIVE Rodents acquire food information from their conspecifics and display a preference for the conspecifics' consumed food. This social learning of food information from others promotes the survival of a species, and it is introduced as the socially transmitted food preference (STFP) task. The cholinergic system in the basal forebrain plays a role in the acquisition of STFP. Cannabidiol (CBD), one of the most abundant phytocannabinoids, exerts its therapeutic potential for cognitive deficits through versatile mechanisms of action, including its interaction with the cholinergic system. We hypothesize a positive relationship between CBD and STFP because acetylcholine (ACh) is involved in STFP, and CBD increases the ACh levels in the basal forebrain. MATERIALS AND METHODS Male C57BL/6J mice were trained to acquire the STFP task. We examined whether CBD affects STFP memory by administering CBD (20 mg/kg, i.p.) before the STFP social training. The involvement of cholinergic system in CBD's effect on STFP was examined by knockdown of brain acetylcholinesterase (AChE), applying a nonselective muscarinic antagonist SCO (3 mg/kg, i.p.) before CBD treatment, and measuring the basal forebrain ACh levels in the CBD-treated mice. RESULTS We first showed that CBD enhanced STFP memory. Knockdown of brain AChE also enhanced STFP memory, which mimicked CBD's effect on STFP. SCO blocked CBD's memory-enhancing effect on STFP. Our most significant finding is that the basal forebrain ACh levels in the CBD-treated mice, but not their control counterparts, were positively correlated with mice's STFP memory performance. CONCLUSION This study indicates that CBD enhances STFP memory in mice. Specifically, those which respond to CBD by increasing the muscarinic-mediated ACh signaling perform better in their STFP memory.
Collapse
Affiliation(s)
- Chih-Yu Chang
- Cannabinoid Signaling Laboratory, Department of Psychology, National Cheng Kung University, 1 University Rd, Tainan, 70101, Taiwan
| | - Wen Dai
- Cannabinoid Signaling Laboratory, Department of Psychology, National Cheng Kung University, 1 University Rd, Tainan, 70101, Taiwan
| | - Sherry Shu-Jung Hu
- Cannabinoid Signaling Laboratory, Department of Psychology, National Cheng Kung University, 1 University Rd, Tainan, 70101, Taiwan.
| |
Collapse
|
10
|
Packer CH, Jasset O, Hanniford N, Brigida S, Demidkin S, Perlis RH, Edlow AG, Shook LL. Maternal-fetal cytokine profiles in acute SARS-CoV-2 "breakthrough" infection after COVID-19 vaccination. Front Immunol 2025; 15:1506203. [PMID: 39845965 PMCID: PMC11750656 DOI: 10.3389/fimmu.2024.1506203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/16/2024] [Indexed: 01/24/2025] Open
Abstract
Objective Vaccination is protective against severe COVID-19 disease, yet whether vaccination reduces COVID-19-associated inflammation in pregnancy has not been established. The objective of this study is to characterize maternal and cord cytokine profiles of acute SARS-CoV-2 "breakthrough" infection (BTI) after vaccination, compared with unvaccinated infection and uninfected controls. Study design 66 pregnant individuals enrolled in the MGH COVID-19 biorepository (March 2020-April 2022) were included. Maternal sera were collected from 26 unvaccinated and 21 vaccinated individuals with acute SARS-CoV-2 infection. Cord sera were collected at delivery. Maternal and cord sera from 19 term dyads without current or prior SARS-CoV-2 infection were analyzed as controls. Cytokines were quantified using the Human Inflammation 20-Plex ProcartaPlex assay. Results There was a significantly higher incidence of severe/critical maternal illness in unvaccinated pregnant individuals with SARS-CoV-2 compared to vaccinated (10/26 (38%) vs. 0/21 (0%), p<0.01). Significantly higher maternal levels of TNFα and CD62P were observed in vaccinated individuals with SARS-CoV-2 BTI compared with unvaccinated individuals with infection (p<0.05). Network correlation analyses revealed a distinct maternal cytokine response to SARS-CoV-2 in vaccinated vs unvaccinated individuals. Neither unvaccinated nor vaccinated SARS-CoV-2 infection resulted in elevated cord cytokines compared to controls. Multivariate analyses demonstrate distinct maternal and cord cytokine profiles in the setting of maternal SARS-CoV-2 at delivery. Conclusion Vaccination was associated with higher maternal cytokine levels during acute SARS-CoV-2 infection compared to unvaccinated infection, which may reflect vaccine-mediated priming of the immune system. A fetal inflammatory response specific to maternal SARS-CoV-2 infection was not observed.
Collapse
Affiliation(s)
- Claire H. Packer
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Olyvia Jasset
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Nikolina Hanniford
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Sara Brigida
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Stepan Demidkin
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Roy H. Perlis
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Andrea G. Edlow
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA, United States
| | - Lydia L. Shook
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
11
|
Otero AM, Connolly MG, Gonzalez-Ricon RJ, Wang SS, Allen JM, Antonson AM. Influenza A virus during pregnancy disrupts maternal intestinal immunity and fetal cortical development in a dose- and time-dependent manner. Mol Psychiatry 2025; 30:13-28. [PMID: 38961232 PMCID: PMC11649561 DOI: 10.1038/s41380-024-02648-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/05/2024]
Abstract
Epidemiological studies link exposure to viral infection during pregnancy, including influenza A virus (IAV) infection, with increased incidence of neurodevelopmental disorders (NDDs) in offspring. Models of maternal immune activation (MIA) using viral mimetics demonstrate that activation of maternal intestinal T helper 17 (TH17) cells, which produce effector cytokine interleukin (IL)-17, leads to aberrant fetal brain development, such as neocortical malformations. Fetal microglia and border-associated macrophages (BAMs) also serve as potential cellular mediators of MIA-induced cortical abnormalities. However, neither the inflammation-induced TH17 cell pathway nor fetal brain-resident macrophages have been thoroughly examined in models of live viral infection during pregnancy. Here, we inoculated pregnant mice with two infectious doses of IAV and evaluated peak innate and adaptive immune responses in the dam and fetus. While respiratory IAV infection led to dose-dependent maternal colonic shortening and microbial dysregulation, there was no elevation in intestinal TH17 cells nor IL-17. Systemically, IAV resulted in consistent dose- and time-dependent increases in IL-6 and IFN-γ. Fetal cortical abnormalities and global changes in fetal brain transcripts were observable in the high-but not the moderate-dose IAV group. Profiling of fetal microglia and BAMs revealed dose- and time-dependent differences in the numbers of meningeal but not choroid plexus BAMs, while microglial numbers and proliferative capacity of Iba1+ cells remained constant. Fetal brain-resident macrophages increased phagocytic CD68 expression, also in a dose- and time-dependent fashion. Taken together, our findings indicate that certain features of MIA are conserved between mimetic and live virus models, while others are not. Overall, we provide consistent evidence of an infection severity threshold for downstream maternal inflammation and fetal cortical abnormalities, which recapitulates a key feature of the epidemiological data and further underscores the importance of using live pathogens in NDD modeling to better evaluate the complete immune response and to improve translation to the clinic.
Collapse
Affiliation(s)
- Ashley M Otero
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Meghan G Connolly
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | | | - Selena S Wang
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jacob M Allen
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Adrienne M Antonson
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
12
|
Mattei D, Guneykaya D, Ugursu B, Buonfiglioli A. From womb to world: The interplay between maternal immune activation, neuroglia, and neurodevelopment. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:269-285. [PMID: 40148048 DOI: 10.1016/b978-0-443-19102-2.00028-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
This chapter introduces and discusses maternal immune activation (MIA) as a contributing factor in increasing the risk of neurodevelopmental disorders, particularly in relation to its interactions with neuroglia. Here we first provide an overview of the neuroglia-astroglia, oligodendroglia, microglia, and radial glial cells-and their important role during early brain development and in adulthood. We then present and discuss MIA, followed by a critical overview of inflammatory molecules and temporal stages associated to maternal inflammation during pregnancy. We provide an overview of animal and human models used to mimic and study MIA. Furthermore, we review the possible interaction between MIA and neuroglia, focusing on the current advances in both modeling and therapeutics. Additionally, we discuss and provide preliminary and interesting insights into the most recent pandemic, COVID-19, and how the infection may be associated to MIA and increased risk for neurodevelopmental disorders. Finally, we provide a critical overview of challenges and future opportunities to study how MIA may contribute to higher risk of developing neurodevelopmental disorders.
Collapse
Affiliation(s)
- Daniele Mattei
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, New York, NY, United States
| | - Dilansu Guneykaya
- Department of Neurobiology, Harvard Medical School, Boston, MA, United States
| | - Bilge Ugursu
- Department of Psychoneuroimmunology, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Alice Buonfiglioli
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
13
|
Prasad J, Van Steenwinckel J, Gunn AJ, Bennet L, Korzeniewski SJ, Gressens P, Dean JM. Chronic Inflammation Offers Hints About Viable Therapeutic Targets for Preeclampsia and Potentially Related Offspring Sequelae. Int J Mol Sci 2024; 25:12999. [PMID: 39684715 PMCID: PMC11640791 DOI: 10.3390/ijms252312999] [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: 08/29/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
The combination of hypertension with systemic inflammation during pregnancy is a hallmark of preeclampsia, but both processes also convey dynamic information about its antecedents and correlates (e.g., fetal growth restriction) and potentially related offspring sequelae. Causal inferences are further complicated by the increasingly frequent overlap of preeclampsia, fetal growth restriction, and multiple indicators of acute and chronic inflammation, with decreased gestational length and its correlates (e.g., social vulnerability). This complexity prompted our group to summarize information from mechanistic studies, integrated with key clinical evidence, to discuss the possibility that sustained or intermittent systemic inflammation-related phenomena offer hints about viable therapeutic targets, not only for the prevention of preeclampsia, but also the neurobehavioral and other developmental deficits that appear to be overrepresented in surviving offspring. Importantly, we feel that carefully designed hypothesis-driven observational studies are necessary if we are to translate the mechanistic evidence into child health benefits, namely because multiple pregnancy disorders might contribute to heightened risks of neuroinflammation, arrested brain development, or dysconnectivity in survivors who exhibit developmental problems later in life.
Collapse
Affiliation(s)
- Jaya Prasad
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| | | | - Alistair J. Gunn
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| | - Laura Bennet
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| | - Steven J. Korzeniewski
- C.S. Mott Center for Human Growth and Development, Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Pierre Gressens
- Inserm, Neurodiderot, Université de Paris, 75019 Paris, France;
- Centre for the Developing Brain, Division of Imaging Sciences and Department of Biomedical Engineering, King’s College London, King’s Health Partners, St. Thomas’ Hospital, London SE1 7EH, UK
| | - Justin M. Dean
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| |
Collapse
|
14
|
Chen T, Meng H, Fang N, Shi P, Chen M, Liu Q, Lv L, Li W. Age-related changes in behavior profile in male offspring of rats treated with poly I:C-induced maternal immune activation in early gestation. Animal Model Exp Med 2024; 7:914-925. [PMID: 38741390 PMCID: PMC11680485 DOI: 10.1002/ame2.12417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 03/21/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Autism and schizophrenia are environmental risk factors associated with prenatal viral infection during pregnancy. It is still unclear whether behavior phenotypes change at different developmental stages in offspring following the activation of the maternal immune system. METHODS Sprague-Dawley rats received a single caudal vein injection of 10 mg/kg polyinosinic:polycytidylic acid (poly I:C) on gestational day 9 and the offspring were comprehensively tested for behaviors in adolescence and adulthood. RESULTS Maternal serum levels of interleukin (IL)-6, IL-1β and tumor necrosis factor-α were elevated in poly I:C-treated dams. The offspring of maternal poly I:C-induced rats showed increased anxiety, impaired social approach, and progressive impaired cognitive and sensorimotor gating function. CONCLUSION Maternal immune activation led to developmental specificity behavioral impairment in offspring.
Collapse
Affiliation(s)
- Tengfei Chen
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical UniversityXinxiangChina
- International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiangChina
| | - Huadan Meng
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical UniversityXinxiangChina
- International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiangChina
| | - Ni Fang
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical UniversityXinxiangChina
- International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiangChina
| | - Peiling Shi
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical UniversityXinxiangChina
- International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiangChina
| | - Mengxue Chen
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical UniversityXinxiangChina
- International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiangChina
| | - Qing Liu
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical UniversityXinxiangChina
- International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiangChina
| | - Luxian Lv
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical UniversityXinxiangChina
- International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiangChina
- Henan Province People's HospitalZhengzhouHenanChina
| | - Wenqiang Li
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical UniversityXinxiangChina
- International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiangChina
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental DisorderXinxiang Medical UniversityXinxiangChina
| |
Collapse
|
15
|
Olasunkanmi OI, Aremu J, Wong ML, Licinio J, Zheng P. Maternal gut-microbiota impacts the influence of intrauterine environmental stressors on the modulation of human cognitive development and behavior. J Psychiatr Res 2024; 180:307-326. [PMID: 39488009 DOI: 10.1016/j.jpsychires.2024.10.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/01/2023] [Accepted: 10/23/2024] [Indexed: 11/04/2024]
Abstract
This review examines the longstanding debate of nature and intrauterine environmental challenges that shapes human development and behavior, with a special focus on the influence of maternal prenatal gut microbes. Recent research has revealed the critical role of the gut microbiome in human neurodevelopment, and evidence suggest that maternal microbiota can impact fetal gene and microenvironment composition, as well as immunophysiology and neurochemical responses. Furthermore, intrauterine neuroepigenetic regulation may be influenced by maternal microbiota, capable of having long-lasting effects on offspring behavior and cognition. By examining the complex relationship between maternal prenatal gut microbes and human development, this review highlights the importance of early-life environmental factors in shaping neurodevelopment and cognition.
Collapse
Affiliation(s)
- Oluwatayo Israel Olasunkanmi
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education) Chongqing Medical University, Chongqing, China.
| | - John Aremu
- Department of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Ma-Li Wong
- Department of Psychiatry, College of Medicine, Upstate Medical University, Syracuse, NY, USA
| | - Julio Licinio
- Department of Psychiatry, College of Medicine, Upstate Medical University, Syracuse, NY, USA.
| | - Peng Zheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education) Chongqing Medical University, Chongqing, China.
| |
Collapse
|
16
|
Wang K, Zhang S, Wang Y, Wu X, Wen L, Meng T, Jin X, Li S, Hong Y, Ke J, Xu Y, Yuan H, Hu F. Taprenepag restores maternal-fetal interface homeostasis for the treatment of neurodevelopmental disorders. J Neuroinflammation 2024; 21:307. [PMID: 39609821 PMCID: PMC11603931 DOI: 10.1186/s12974-024-03300-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 11/16/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND AND PURPOSE Neurodevelopmental disorders (NDDs) are characterized by abnormalities in brain development and neurobehaviors, including autism. The maternal-fetal interface (MFI) is a highly specialized tissue through which maternal factors affect fetal brain development. However, limited research exists on restoring and maintaining MFI homeostasis and its potential impact on NDDs. This study explores the role of placental indoleamine 2,3-dioxygenase (IDO-1) in MFI homeostasis and fetal brain development. EXPERIMENTAL APPROACH The maternal-fetal barrier was disrupted by sodium valproate (VPA) in pregnant mice, whose offspring show typical autism-like behaviors. Ultrastructural analysis and flow cytometric analysis were conducted to observe the morphological and immune system changes. Behavioral tests and immunofluorescence staining was used to investigate the ability and mechanism of taprenepag to alleviate the abnormal behaviors of VPA-exposed offspring and normalize the development of serotonergic neurons. KEY RESULTS In VPA-exposed pregnant mice, the downregulation of IDO-1 led to maternal immune overactivation and disruption of maternal-fetal barrier, resulting in excessive 5-HT synthesis in the placenta. This process disrupted the development of the serotonergic neuronal system in the offspring, resulting in impaired development of serotonergic neurons, thalamocortical axons, and NDDs in the progeny. However, a single injection of taprenepag at E13.5 ultimately upregulated placental IDO-1 through amplifying the positive feedback loop COX-2/PGE2/PTGER-2/IDO-1 and abolished these alterations. CONCLUSION Taprenepag improved autism-like behaviors in the offspring of VPA-exposed mice by addressing placental IDO-1 downregulation. This study highlights the potential of targeting IDO-1 to mitigate MFI disruption and NDD development.
Collapse
Affiliation(s)
- Kai Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, PR China
| | - Shufen Zhang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Yunxia Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Xiaomei Wu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Lijuan Wen
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, PR China
| | - Tingting Meng
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, PR China
| | - Xiangyu Jin
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, PR China
| | - Sufen Li
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Yiling Hong
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Jia Ke
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Yichong Xu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Hong Yuan
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, PR China
| | - Fuqiang Hu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
- Jinhua Institute of Zhejiang University, Jinhua, 321299, PR China.
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, College of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China.
| |
Collapse
|
17
|
Wang K, Liu S, Huang D, Guan X, Chen N, Xiu M, Liu D, Huang Y. Onset age moderates the associations between neutrophil-to-lymphocyte ratio and clinical symptoms in first-episode patients with schizophrenia. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2024; 10:110. [PMID: 39562579 DOI: 10.1038/s41537-024-00522-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 09/27/2024] [Indexed: 11/21/2024]
Abstract
Patients with schizophrenia with early onset age have been shown to exhibit more severe negative symptoms. Genetic, biomarker, postmortem brain, and imaging studies indicate the involvement of immune abnormalities in the pathophysiology of schizophrenia. In this study, we examined the moderating role of early onset on the associations between clinical symptoms and neutrophil-to-lymphocyte ratio (NLR) in medication-naïve first-episode schizophrenia (MNFES). A total of 97 MNFES patients were recruited. Neutrophil (NEU), LYM, and NLR values were compared between early-onset (EO) and non-early-onset (non-EO) patients with schizophrenia to explore the potential influence of EO on the correlations between NLR and symptoms. The results showed no differences in NEU and NLR values between the EO and non-EO groups. In the EO group, NEU and NLR values significantly correlated with general psychopathology and total score (all p < 0.05), whereas lymphocyte counts were not correlated with symptoms of schizophrenia. NEU and NLR were not associated with symptoms in the non-EO group. Linear regression analysis in the EO group revealed that NEU or NLR values were a predictive biomarker for the clinical symptoms. Our study indicates that EO patients had greater severe negative symptoms compared with non-EO patients. In addition, onset age mediates the relationships of NEU and NLR values with clinical symptoms, suggesting that an immune disturbance, particularly increased innate immune response in EO patients, may be involved in the psychophysiology of schizophrenia.
Collapse
Affiliation(s)
- Kuiyuan Wang
- Ganzhou City Key Laboratory of Mental Health, The Third People's Hospital of Ganzhou City, Ganzhou, China
| | - Shaohua Liu
- Ganzhou City Key Laboratory of Mental Health, The Third People's Hospital of Ganzhou City, Ganzhou, China
| | - Dan Huang
- Ganzhou City Key Laboratory of Mental Health, The Third People's Hospital of Ganzhou City, Ganzhou, China
| | - Xiaoni Guan
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, China
| | - Nan Chen
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, China
| | - Meihong Xiu
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, China.
| | - Dianying Liu
- Ganzhou City Key Laboratory of Mental Health, The Third People's Hospital of Ganzhou City, Ganzhou, China.
| | - Yuanyuan Huang
- Department of Psychiatry, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China.
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China.
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
18
|
Collins B, Lemanski EA, Wright-Jin E. The Importance of Including Maternal Immune Activation in Animal Models of Hypoxic-Ischemic Encephalopathy. Biomedicines 2024; 12:2559. [PMID: 39595123 PMCID: PMC11591850 DOI: 10.3390/biomedicines12112559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a perinatal brain injury that is the leading cause of cerebral palsy, developmental delay, and poor cognitive outcomes in children born at term, occurring in about 1.5 out of 1000 births. The only proven therapy for HIE is therapeutic hypothermia. However, despite this treatment, many children ultimately suffer disability, brain injury, and even death. Barriers to implementation including late diagnosis and lack of resources also lead to poorer outcomes. This demonstrates a critical need for additional treatments for HIE, and to facilitate this, we need translational models that accurately reflect risk factors and interactions present in HIE. Maternal or amniotic infection is a significant risk factor and possible cause of HIE in humans. Maternal immune activation (MIA) is a well-established model of maternal infection and inflammation that has significant developmental consequences largely characterized within the context of neurodevelopmental disorders such as autism spectrum disorder and schizophrenia. MIA can also lead to long-lasting changes within the neuroimmune system, which lead to compounding negative outcomes following a second insult. This supports the importance of understanding the interaction of maternal inflammation and hypoxic-ischemic outcomes. Animal models have been invaluable to understanding the pathophysiology of this injury and to the development of therapeutic hypothermia. However, each model system has its own limitations. Large animal models such as pigs may more accurately represent the brain and organ development and complexity in humans, while rodent models are more cost-effective and offer more possible molecular techniques. Recent studies have utilized MIA or direct inflammation prior to HIE insult. Investigators should thoughtfully consider the risk factors they wish to include in their HIE animal models. In the incorporation of MIA, investigators should consider the type, timing, and dose of the inflammatory stimulus, as well as the timing, severity, and type of hypoxic insult. Using a variety of animal models that incorporate the maternal-placental-fetal system of inflammation will most likely lead to a more robust understanding of the mechanisms of this injury that can guide future clinical decisions and therapies.
Collapse
Affiliation(s)
- Bailey Collins
- Division of Biomedical Research, Nemours Children’s Health, Wilmington, DE 19803, USA; (B.C.); (E.A.L.)
- Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA
| | - Elise A. Lemanski
- Division of Biomedical Research, Nemours Children’s Health, Wilmington, DE 19803, USA; (B.C.); (E.A.L.)
- Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA
| | - Elizabeth Wright-Jin
- Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA
- Division of Neurology, Nemours Children’s Health, Wilmington, DE 19803, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
19
|
Zeng X, Fan L, Qin Q, Zheng D, Wang H, Li M, Jiang Y, Wang H, Liu H, Liang S, Wu L, Liang S. Exogenous PD-L1 binds to PD-1 to alleviate and prevent autism-like behaviors in maternal immune activation-induced male offspring mice. Brain Behav Immun 2024; 122:527-546. [PMID: 39182588 DOI: 10.1016/j.bbi.2024.08.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024] Open
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder caused by the interaction of multiple pathogenic factors. Epidemiological studies and animal experiments indicate that maternal immune activation (MIA) is closely related to the development of ASD in offspring. A large number of pro-inflammatory cytokines are transferred from the placenta to the fetal brain during MIA, which impedes fetal neurodevelopment and is accompanied by activation of immune cells and microglia. Programmed cell death protein 1 (PD-1) can be highly expressed on the surface of various activated immune cells, when combined with programmed cell death-ligand 1 (PD-L1), it can activate the PD-1/PD-L1 pathway and exert powerful immunosuppressive effects, suggesting that this immune checkpoint may have the potential to treat MIA-induced ASD. This study combined bioinformatics analysis and experimental validation to explore the efficacy of Fc-fused PD-L1 (PD-L1-Fc) in treating MIA-induced ASD. Bioinformatics analysis results showed that in human placental inflammation, IL-6 was upregulated, T cells proliferated significantly, and the PD-1/PD-L1 pathway was significantly enriched. The experimental results showed that intraperitoneal injection of poly(I:C) induced MIA in pregnant mice resulted in significant expression of IL-6 in their serum, placenta, and fetal brain. At the same time, the expression of PD-1 and PD-L1 in the placenta and fetal brain increased, CD4+ T cells in the spleen were significantly activated, and PD-1 expression increased. Their offspring mice exhibited typical ASD-like behaviors. In vitro experiments on primary microglia of offspring mice have confirmed that the expression of IL-6, PD-1, and PD-L1 is significantly increased, and PD-L1-Fc effectively reduced their expression levels. In the prefrontal cortex of MIA offspring mice, there was an increase in the expression of IL-6, PD-1, and PD-L1; activation of microglial cells, and colocalization with PD-1. Then we administered brain stereotaxic injections of PD-L1-Fc to MIA offspring mice and intraperitoneal injections to MIA pregnant mice. The results indicated that PD-L1-Fc effectively suppressed neuroinflammation in the frontal cortex of offspring mice and partially ameliorated ASD-like behaviors; MIA in pregnant mice was significantly alleviated, and the offspring mice they produced did not exhibit neuroinflammation or ASD-like behaviors. In summary, we have demonstrated the therapeutic ability of PD-L1-Fc for MIA-induced ASD, aiming to provide new strategies and insights for the treatment of ASD.
Collapse
Affiliation(s)
- Xin Zeng
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Linlin Fan
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Qian Qin
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Danyang Zheng
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Han Wang
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Mengyue Li
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Yutong Jiang
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Hui Wang
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Hao Liu
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Shengjun Liang
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Lijie Wu
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China.
| | - Shuang Liang
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
20
|
Jaswa EG, Huddleston HG, Lindquist KJ, Wu AHB, Bishop SL, Kim YS, Kaing A, Prahl M, Gaw SL, Corley J, Hoskin E, Cho YJ, Rogers EE, Cedars MI. In Utero Exposure to Maternal COVID-19 and Offspring Neurodevelopment Through Age 24 Months. JAMA Netw Open 2024; 7:e2439792. [PMID: 39412802 PMCID: PMC11581627 DOI: 10.1001/jamanetworkopen.2024.39792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 08/18/2024] [Indexed: 11/24/2024] Open
Abstract
Importance In utero exposure to maternal infections has been associated with abnormal neurodevelopment among offspring. The emergence of a new, now endemic infection (SARS-CoV-2) warrants investigating developmental implications for exposed offspring. Objective To assess whether in utero exposure to maternal COVID-19 is associated with abnormal neurodevelopmental scores among children ages 12, 18, and 24 months. Design, Setting, and Participants Data were ascertained from the ASPIRE (Assessing the Safety of Pregnancy in the Coronavirus Pandemic) trial, a prospective cohort of pregnant individuals aged 18 years or older who were enrolled before 10 weeks' gestation and their children. Individuals were recruited online from May 14, 2020, to August 23, 2021, using the Society for Assisted Reproductive Technology and BabyCenter, an online media platform. Participants from all 50 states and Puerto Rico completed activities remotely. Exposure In utero exposure to COVID-19. Main Outcomes and Measures Birth mothers completed the Ages & Stages Questionnaires, Third Edition, a validated screening tool for developmental delays, at 12, 18, and 24 months' post partum. A score below the cutoff in any domain (communication, gross motor, fine motor, problem-solving, and social skills) was considered an abnormal developmental screen (scores range from 0 to 60 in each domain, with higher scores indicating less risk for neurodevelopmental delay). Results The cohort included 2003 pregnant individuals (mean [SD] age, 33.3 [4.2] years) enrolled before 10 weeks' gestation and who completed study activities; 1750 (87.4%) had earned a college degree. Neurodevelopmental outcomes were available for 1757 children at age 12 months, 1522 at age 18 months, and 1523 at age 24 months. The prevalence of abnormal screens for exposed vs unexposed offspring at age 12 months was 64 of 198 (32.3%) vs 458 of 1559 (29.4%); at age 18 months, 36 of 161 (22.4%) vs 279 of 1361 (20.5%); and at age 24 months, 29 of 151 (19.2%) vs 230 of 1372 (16.8%). In an adjusted mixed-effects logistics regression model, no difference in risk of abnormal neurodevelopmental screens was observed at age 12 months (adjusted risk ratio [ARR], 1.07 [95% CI, 0.85-1.34]), age 18 months (ARR, 1.15 [95% CI, 0.84-1.57]), or age 24 months (ARR, 1.01 [95% CI, 0.69-1.48]). Supplemental analyses did not identify differential risk based on trimester of infection, presence vs absence of fever, or breakthrough infection following vaccination vs primary infection. Conclusions and Relevance In this cohort study of pregnant individuals and offspring, exposure to maternal COVID-19 was not associated with abnormal neurodevelopmental screening results through 24 months' post partum. Continued study of diverse groups of children is needed because, among other factors, evidence suggests sensitivity of the developing fetal brain to maternal immune activation.
Collapse
Affiliation(s)
- Eleni G. Jaswa
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco
| | - Heather G. Huddleston
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco
| | - Karla J. Lindquist
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Alan H. B. Wu
- Division of Clinical Chemistry, Department of Laboratory Medicine, University of California, San Francisco
| | - Somer L. Bishop
- Department of Psychiatry, University of California, San Francisco
| | - Young-Shin Kim
- Department of Psychiatry, University of California, San Francisco
| | - Amy Kaing
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco
| | - Mary Prahl
- Division of Pediatric Infectious Disease and Global Health, Department of Pediatrics, University of California, San Francisco
| | - Stephanie L. Gaw
- Division of Maternal Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco
| | - Jamie Corley
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco
| | - Elena Hoskin
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco
| | - Yoon Jae Cho
- Department of Psychiatry, University of California, San Francisco
| | - Elizabeth E. Rogers
- Division of Neonatology, Department of Pediatrics, University of California, San Francisco
| | - Marcelle I. Cedars
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco
| |
Collapse
|
21
|
Tagliatti E, Bizzotto M, Morini R, Filipello F, Rasile M, Matteoli M. Prenatal drivers of microglia vulnerability in the adult. Immunol Rev 2024; 327:100-110. [PMID: 39508795 DOI: 10.1111/imr.13418] [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] [Indexed: 11/15/2024]
Abstract
Environmental insults during early development heavily affect brain trajectories. Among these, maternal infections, high-fat diet regimens, and sleep disturbances pose a significant risk for neurodevelopmental derangements in the offspring. Notably, scattered evidence is starting to emerge that also paternal lifestyle habits may impact the offspring development. Given their key role in controlling neurogenesis, synaptogenesis and shaping neuronal circuits, microglia represent the most likely suspects of mediating the detrimental effects of prenatal insults. For some of these environmental triggers, like maternal infections, ample literature evidence demonstrates the central role of microglia, also delineating the specific transcriptomic and proteomic profiles induced by these insults. In other contexts, the analysis of microglia is still in its infancy. Fostering these studies is needed to define microglia as potential therapeutic target in the frame of disorders consequent to maternal immune activation.
Collapse
Affiliation(s)
| | | | | | | | - Marco Rasile
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Michela Matteoli
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| |
Collapse
|
22
|
ElGrawani W, Mueller FS, Schalbetter SM, Brown SA, Weber-Stadlbauer U, Tarokh L. Maternal immune activation exerts long-term effects on activity and sleep in male offspring mice. Eur J Neurosci 2024; 60:5505-5521. [PMID: 39210746 DOI: 10.1111/ejn.16506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/16/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
Exposure to infectious or non-infectious immune activation during early development is a serious risk factor for long-term behavioural dysfunctions. Mouse models of maternal immune activation (MIA) have increasingly been used to address neuronal and behavioural dysfunctions in response to prenatal infections. One commonly employed MIA model involves administering poly(I:C) (polyriboinosinic-polyribocytdilic acid), a synthetic analogue of double-stranded RNA, during gestation, which robustly induces an acute viral-like inflammatory response. Using electroencephalography (EEG) and infrared (IR) activity recordings, we explored alterations in sleep/wake, circadian and locomotor activity patterns on the adult male offspring of poly(I:C)-treated mothers. Our findings demonstrate that these offspring displayed reduced home cage activity during the (subjective) night under both light/dark or constant darkness conditions. In line with this finding, these mice exhibited an increase in non-rapid eye movement (NREM) sleep duration as well as an increase in sleep spindles density. Following sleep deprivation, poly(I:C)-exposed offspring extended NREM sleep duration and prolonged NREM sleep bouts during the dark phase as compared with non-exposed mice. Additionally, these mice exhibited a significant alteration in NREM sleep EEG spectral power under heightened sleep pressure. Together, our study highlights the lasting effects of infection and/or immune activation during pregnancy on circadian activity and sleep/wake patterns in the offspring.
Collapse
Affiliation(s)
- Waleed ElGrawani
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Flavia S Mueller
- Institute of Pharmacology and Toxicology, University of Zurich - Vetsuisse, Zurich, Switzerland
| | - Sina M Schalbetter
- Institute of Pharmacology and Toxicology, University of Zurich - Vetsuisse, Zurich, Switzerland
| | - Steven A Brown
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Ulrike Weber-Stadlbauer
- Institute of Pharmacology and Toxicology, University of Zurich - Vetsuisse, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Leila Tarokh
- Translational Research Center, University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
- University Hospital of Child and Adolescent Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| |
Collapse
|
23
|
Maddock RJ, Vlasova RM, Chen S, Iosif AM, Bennett J, Tanase C, Ryan AM, Murai T, Hogrefe CE, Schumann CD, Geschwind DH, Van de Water J, Amaral DG, Lesh TA, Styner MA, Kimberley McAllister A, Carter CS, Bauman MD. Altered brain metabolites in male nonhuman primate offspring exposed to maternal immune activation. Brain Behav Immun 2024; 121:280-290. [PMID: 39032543 PMCID: PMC11809764 DOI: 10.1016/j.bbi.2024.07.011] [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: 03/21/2024] [Revised: 07/04/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024] Open
Abstract
Converging data show that exposure to maternal immune activation (MIA) in utero alters brain development in animals and increases the risk of neurodevelopmental disorders in humans. A recently developed non-human primate MIA model affords opportunities for studies with uniquely strong translational relevance to human neurodevelopment. The current longitudinal study used 1H-MRS to investigate the developmental trajectory of prefrontal cortex metabolites in male rhesus monkey offspring of dams (n = 14) exposed to a modified form of the inflammatory viral mimic, polyinosinic:polycytidylic acid (Poly IC), in the late first trimester. Brain metabolites in these animals were compared to offspring of dams that received saline (n = 10) or no injection (n = 4). N-acetylaspartate (NAA), glutamate, creatine, choline, myo-inositol, taurine, and glutathione were estimated from PRESS and MEGA-PRESS acquisitions obtained at 6, 12, 24, 36, and 45 months of age. Prior investigations of this cohort reported reduced frontal cortical gray and white matter and subtle cognitive impairments in MIA offspring. We hypothesized that the MIA-induced neurodevelopmental changes would extend to abnormal brain metabolite levels, which would be associated with the observed cognitive impairments. Prefrontal NAA was significantly higher in the MIA offspring across all ages (p < 0.001) and was associated with better performance on the two cognitive measures most sensitive to impairment in the MIA animals (both p < 0.05). Myo-inositol was significantly lower across all ages in MIA offspring but was not associated with cognitive performance. Taurine was elevated in MIA offspring at 36 and 45 months. Glutathione did not differ between groups. MIA exposure in male non-human primates is associated with altered prefrontal cortex metabolites during childhood and adolescence. A positive association between elevated NAA and cognitive performance suggests the hypothesis that elevated NAA throughout these developmental stages reflects a protective or resilience-related process in MIA-exposed offspring. The potential relevance of these findings to human neurodevelopmental disorders is discussed.
Collapse
Affiliation(s)
- Richard J Maddock
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California Davis, Sacramento, CA, USA.
| | - Roza M Vlasova
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Shuai Chen
- Division of Biostatistics, Department of Public Health Sciences, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Ana-Maria Iosif
- Division of Biostatistics, Department of Public Health Sciences, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Jeffrey Bennett
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Costin Tanase
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Amy M Ryan
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Takeshi Murai
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Casey E Hogrefe
- California National Primate Research Center, University of California Davis, Davis, CA, USA
| | - Cynthia D Schumann
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Daniel H Geschwind
- Neurogenetics Program, Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Judy Van de Water
- Rheumatology/Allergy and Clinical Immunology, School of Medicine, University of California Davis, Sacramento, CA, USA; MIND Institute, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - David G Amaral
- MIND Institute, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Tyler A Lesh
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Martin A Styner
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA; Department of Computer Science, University of North Carolina, Chapel Hill, NC, USA
| | | | - Cameron S Carter
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California Davis, Sacramento, CA, USA.
| | - Melissa D Bauman
- California National Primate Research Center, University of California Davis, Davis, CA, USA; MIND Institute, School of Medicine, University of California Davis, Sacramento, CA, USA; Physiology and Membrane Biology, School of Medicine, University of California Davis, Sacramento, CA, USA.
| |
Collapse
|
24
|
Wilson JD, Dworsky-Fried M, Ismail N. Neurodevelopmental implications of COVID-19-induced gut microbiome dysbiosis in pregnant women. J Reprod Immunol 2024; 165:104300. [PMID: 39004033 DOI: 10.1016/j.jri.2024.104300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/25/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024]
Abstract
The global public health emergency of COVID-19 in January 2020 prompted a surge in research focusing on the pathogenesis and clinical manifestations of the virus. While numerous reports have been published on the acute effects of COVID-19 infection, the review explores the multifaceted long-term implications of COVID-19, with a particular focus on severe maternal COVID-19 infection, gut microbiome dysbiosis, and neurodevelopmental disorders in offspring. Severe COVID-19 infection has been associated with heightened immune system activation and gastrointestinal symptoms. Severe COVID-19 may also result in gut microbiome dysbiosis and a compromised intestinal mucosal barrier, often referred to as 'leaky gut'. Increased gut permeability facilitates the passage of inflammatory cytokines, originating from the inflamed intestinal mucosa and gut, into the bloodstream, thereby influencing fetal development during pregnancy and potentially elevating the risk of neurodevelopmental disorders such as autism and schizophrenia. The current review discusses the role of cytokine signaling molecules, microglia, and synaptic pruning, highlighting their potential involvement in the pathogenesis of neurodevelopmental disorders following maternal COVID-19 infection. Additionally, this review addresses the potential of probiotic interventions to mitigate gut dysbiosis and inflammatory responses associated with COVID-19, offering avenues for future research in optimizing maternal and fetal health outcomes.
Collapse
Affiliation(s)
- Jacob D Wilson
- NISE Laboratory, School of Psychology, Faculty of Social Science, University of Ottawa, Ottawa, Ontario K1N 9A4, Canada
| | - Michaela Dworsky-Fried
- NISE Laboratory, School of Psychology, Faculty of Social Science, University of Ottawa, Ottawa, Ontario K1N 9A4, Canada
| | - Nafissa Ismail
- NISE Laboratory, School of Psychology, Faculty of Social Science, University of Ottawa, Ottawa, Ontario K1N 9A4, Canada; LIFE Research Institute, Ottawa, Ontario K1N 6N5, Canada; University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario K1H 8M5, Canada.
| |
Collapse
|
25
|
Collins JM, Keane JM, Deady C, Khashan AS, McCarthy FP, O'Keeffe GW, Clarke G, Cryan JF, Caputi V, O'Mahony SM. Prenatal stress impacts foetal neurodevelopment: Temporal windows of gestational vulnerability. Neurosci Biobehav Rev 2024; 164:105793. [PMID: 38971516 DOI: 10.1016/j.neubiorev.2024.105793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Prenatal maternal stressors ranging in severity from everyday occurrences/hassles to the experience of traumatic events negatively impact neurodevelopment, increasing the risk for the onset of psychopathology in the offspring. Notably, the timing of prenatal stress exposure plays a critical role in determining the nature and severity of subsequent neurodevelopmental outcomes. In this review, we evaluate the empirical evidence regarding temporal windows of heightened vulnerability to prenatal stress with respect to motor, cognitive, language, and behavioural development in both human and animal studies. We also explore potential temporal windows whereby several mechanisms may mediate prenatal stress-induced neurodevelopmental effects, namely, excessive hypothalamic-pituitary-adrenal axis activity, altered serotonin signalling and sympathetic-adrenal-medullary system, changes in placental function, immune system dysregulation, and alterations of the gut microbiota. While broadly defined developmental windows are apparent for specific psychopathological outcomes, inconsistencies arise when more complex cognitive and behavioural outcomes are considered. Novel approaches to track molecular markers reflective of the underlying aetiologies throughout gestation to identify tractable biomolecular signatures corresponding to critical vulnerability periods are urgently required.
Collapse
Affiliation(s)
- James M Collins
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - James M Keane
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
| | - Clara Deady
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - Ali S Khashan
- School of Public Health, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland.
| | - Fergus P McCarthy
- The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland; Department of Obstetrics and Gynaecology, University College Cork, Cork, Ireland.
| | - Gerard W O'Keeffe
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland.
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland.
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - Valentina Caputi
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | | |
Collapse
|
26
|
Schaer R, Mueller FS, Notter T, Weber-Stadlbauer U, Meyer U. Intrauterine position effects in a mouse model of maternal immune activation. Brain Behav Immun 2024; 120:391-402. [PMID: 38897330 DOI: 10.1016/j.bbi.2024.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 05/27/2024] [Accepted: 06/16/2024] [Indexed: 06/21/2024] Open
Abstract
Rodent models of maternal immune activation (MIA) are increasingly used as experimental tools in preclinical research of immune-mediated neurodevelopmental disorders and mental illnesses. Using a viral-like MIA model that is based on prenatal poly(I:C) exposure in mice, we have recently identified the existence of subgroups of MIA-exposed offspring that show dissociable behavioral, transcriptional, brain network and inflammatory profiles even under conditions of genetic homogeneity and identical MIA. Here, we tested the hypothesis that the intrauterine positions of fetuses, which are known to shape individual variability in litter-bearing mammals through variations in fetal hormone exposure, may contribute to the variable outcomes of MIA in mice. MIA was induced by maternal administration of poly(I:C) on gestation day 12 in C57BL/6N mice. Determining intrauterine positions using delivery by Cesarean section (C-section), we found that MIA-exposed offspring developing between female fetuses only (0M-MIA offspring) displayed significant deficits in sociability and sensorimotor gating at adult age, whereas MIA-exposed offspring developing between one or two males in utero (1/2M-MIA offspring) did not show the same deficits. These intrauterine position effects similarly emerged in male and female offspring. Furthermore, while MIA elevated fetal brain levels of pro- and anti-inflammatory cytokines independently of the precise intrauterine position and sex of adjacent fetuses during the acute phase, fetal brain levels of TNF-α remained elevated in 0M-MIA but not 1/2M-MIA offspring until the post-acute phase in late gestation. As expected, 1/2M offspring generally showed higher testosterone levels in the fetal brain during late gestation as compared to 0M offspring, confirming the transfer of testosterone from male fetuses to adjacent male or female fetuses. Taken together, our findings identify a novel source of within-litter variability contributing to heterogeneous outcomes of short- and long-term effects in a mouse model of MIA. In broader context, our findings highlight that individual differences in fetal exposure to hormonal and inflammatory signals may be a perinatal factor that shapes risk and resilience to MIA.
Collapse
Affiliation(s)
- Ron Schaer
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Flavia S Mueller
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Tina Notter
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Ulrike Weber-Stadlbauer
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Urs Meyer
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
27
|
Mattei D, Ivanov A, Hammer J, Ugursu B, Schalbetter S, Richetto J, Weber-Stadlbauer U, Mueller F, Scarborough J, Wolf SA, Kettenmann H, Wollscheid B, Beule D, Meyer U. Microglia undergo molecular and functional adaptations to dark and light phases in male laboratory mice. Brain Behav Immun 2024; 120:571-583. [PMID: 38986723 DOI: 10.1016/j.bbi.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/20/2024] [Accepted: 07/06/2024] [Indexed: 07/12/2024] Open
Abstract
Microglia are increasingly recognized to contribute to brain health and disease. Preclinical studies using laboratory rodents are essential to advance our understanding of the physiological and pathophysiological roles of these cells in the central nervous system. Rodents are nocturnal animals, and they are mostly maintained in a defined light-dark cycle within animal facilities, with many laboratories investigating the molecular and functional profiles of microglia exclusively during the animals' light (sleep) phase. However, only a few studies have considered possible differences in microglial functions between the active and sleep phases. Based on initial evidence suggesting that microglial intrinsic clock genes can affect their phenotypes, we sought to investigate differences in transcriptional, proteotype and functional profiles of microglia between light (sleep) and dark (active) phases, and how these changes are affected in pathological models. We found marked transcriptional and proteotype differences between microglia harvested from male mice during the light or dark phase. Amongst others, these differences related to genes and proteins associated with immune responses, motility, and phagocytosis, which were reflected by functional alterations in microglial synaptic pruning and response to bacterial stimuli. Possibly accounting for such changes, we found RNA and protein regulation in SWI/SNF and NuRD chromatin remodeling complexes between light and dark phases. Importantly, we also show that the time of microglial sample collection influences the nature of microglial transcriptomic changes in a model of immune-mediated neurodevelopmental disorders. Our findings emphasize the importance of considering diurnal factors in studying microglial cells and indicate that implementing a circadian perspective is pivotal for advancing our understanding of their physiological and pathophysiological roles in brain health and disease.
Collapse
Affiliation(s)
- Daniele Mattei
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland; Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America.
| | - Andranik Ivanov
- Core Unit Bioinformatics, Berlin Institute of Health, Charité-Universitätsmedizin, Berlin, Germany
| | - Jacqueline Hammer
- Institute of Molecular Systems Biology and Department for Health Sciences and Technology, ETH Zürich, Switzerland
| | - Bilge Ugursu
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Department of Ophthalmology, Charité - Universitätsmedizin Berlin, Germany; Psychoneuroimmunology, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Sina Schalbetter
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland
| | - Juliet Richetto
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland
| | - Ulrike Weber-Stadlbauer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland
| | - Flavia Mueller
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland
| | - Joseph Scarborough
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland
| | - Susanne A Wolf
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Department of Ophthalmology, Charité - Universitätsmedizin Berlin, Germany; Psychoneuroimmunology, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Helmut Kettenmann
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Bernd Wollscheid
- Institute of Molecular Systems Biology and Department for Health Sciences and Technology, ETH Zürich, Switzerland
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health, Charité-Universitätsmedizin, Berlin, Germany
| | - Urs Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
28
|
Wu Y, Su Q. Harnessing the Gut Microbiome: To What Extent Can Pre-/Probiotics Alleviate Immune Activation in Autism Spectrum Disorder? Nutrients 2024; 16:2382. [PMID: 39125263 PMCID: PMC11314583 DOI: 10.3390/nu16152382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Children diagnosed with autism spectrum disorder (ASD) are at an increased risk of experiencing gastrointestinal (GI) discomfort, which has been linked to dysfunctions in the microbiome-gut-brain axis. The bidirectional communication between gut and brain plays a crucial role in the overall health of individuals, and alterations in the gut microbiome can contribute to immune activation and gut-brain dysfunction in ASD. Despite the limited and controversial results of pre-/probiotic applications in ASD, this review comprehensively maps the association between ASD clinical symptoms and specific bacterial taxa and evaluates the efficacy of pre-/probiotics in modulating microbiota composition, reducing inflammatory biomarkers, alleviating difficulties in GI distress, sleep problems, core and other ASD-associated symptoms, as well as relieving parental concerns, separately, in individuals with ASD. Beyond simply targeting core ASD symptoms, this review highlights the potential of pre-/probiotic supplementations as a strategy to modulate gut homeostasis and immune response, and to delineate the potential mechanisms by which its direct or mediating effects can alleviate gut-brain dysfunction and poor nutritional status in ASD management. Further well-designed randomized controlled trials are needed to strengthen the existing evidence and establish optimal protocols for the use of pre-/probiotics in the context of ASD.
Collapse
Affiliation(s)
- Yuqi Wu
- Microbiota I-Center (MagIC), Hong Kong SAR, China;
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qi Su
- Microbiota I-Center (MagIC), Hong Kong SAR, China;
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
29
|
Tillmann KE, Schaer R, Mueller FS, Mueller K, Voelkl B, Weber-Stadlbauer U, Pollak DD. Differential effects of purified low molecular weight Poly(I:C) in the maternal immune activation model depend on the laboratory environment. Transl Psychiatry 2024; 14:300. [PMID: 39033141 PMCID: PMC11271296 DOI: 10.1038/s41398-024-03014-7] [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] [Received: 11/13/2023] [Revised: 06/27/2024] [Accepted: 07/04/2024] [Indexed: 07/23/2024] Open
Abstract
The Poly (I:C) (polyriboinosinic-polyribocytidilic acid) paradigm of maternal immune activation (MIA) is most widely used as experimental model for the evaluation of the effects of gestational infection on the brain and behavior of the progeny. We have previously reported significant batch-to-batch variability in the effects of Poly (I:C), purchased from the same supplier (Sigma-Aldrich), on maternal and fetal immune responses and found these differences to be dependent on the relative amount of synthetic double-stranded RNA fragments in the high versus low molecular weight (LMW) range contained in the compound. We here resorted to Poly (I:C) purified for LMW dsRNA fragments to establish a MIA paradigm with increased reproducibility and enhanced standardization in an effort to refine the MIA paradigm and characterize its effect on offspring behavior. We found that the parallel application of LMW Poly (I:C) in two different MIA-experienced laboratories (Vienna and Zurich) yielded differential outcomes in terms of maternal immune responses and behavioral phenotypes in the offspring generation. In both experimental sites, administration of LMW Poly (I:C) induced a significant sickness response and cytokine induction in the pregnant dam and fetal brains, while the expected deficit in sociability as one main behavioral outcome parameter in the MIA progeny, was only present in the Zurich, but not the Vienna cohort. We conclude that although using Poly (I:C) purified for a defined molecular weight range reduces batch-to-batch variability, it does not make the MIA model more reliable and robust. The differential response in behavioral phenotypes of the MIA offspring between the two laboratories illustrates the highly complex interaction between prenatal and postnatal milieus - including the laboratory environment - that determine offspring phenotypic outcomes after MIA. Consequently, establishing a new MIA protocol or implementing the MIA model firstly under new or changed environmental conditions must include the assessment of offspring behavior to ensure solid and reproducible experimental outcomes.
Collapse
Affiliation(s)
- Katharina E Tillmann
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Ron Schaer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Flavia S Mueller
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Karin Mueller
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Bernhard Voelkl
- Animal Welfare Division, Veterinary Public Health Institute University of Bern, Bern, Switzerland
| | - Ulrike Weber-Stadlbauer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland.
- Neuroscience Center Zurich, University of Zurich and ETH, Winterthurerstrasse 190, 8057, Zurich, Switzerland.
| | - Daniela D Pollak
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
30
|
Mastenbroek LJM, Kooistra SM, Eggen BJL, Prins JR. The role of microglia in early neurodevelopment and the effects of maternal immune activation. Semin Immunopathol 2024; 46:1. [PMID: 38990389 PMCID: PMC11239780 DOI: 10.1007/s00281-024-01017-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024]
Abstract
Activation of the maternal immune system during gestation has been associated with an increased risk for neurodevelopmental disorders in the offspring, particularly schizophrenia and autism spectrum disorder. Microglia, the tissue-resident macrophages of the central nervous system, are implicated as potential mediators of this increased risk. Early in development, microglia start populating the embryonic central nervous system and in addition to their traditional role as immune responders under homeostatic conditions, microglia are also intricately involved in various early neurodevelopmental processes. The timing of immune activation may interfere with microglia functioning during early neurodevelopment, potentially leading to long-term consequences in postnatal life. In this review we will discuss the involvement of microglia in brain development during the prenatal and early postnatal stages of life, while also examining the effects of maternal immune activation on microglia and neurodevelopmental processes. Additionally, we discuss recent single cell RNA-sequencing studies focusing on microglia during prenatal development, and hypothesize how early life microglial priming, potentially through epigenetic reprogramming, may be related to neurodevelopmental disorders.
Collapse
Affiliation(s)
- L J M Mastenbroek
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - S M Kooistra
- Department of BioMedical Sciences, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - B J L Eggen
- Department of BioMedical Sciences, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - J R Prins
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
31
|
Zoicas I, Licht C, Mühle C, Kornhuber J. Repetitive transcranial magnetic stimulation (rTMS) for depressive-like symptoms in rodent animal models. Neurosci Biobehav Rev 2024; 162:105726. [PMID: 38762128 DOI: 10.1016/j.neubiorev.2024.105726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/30/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024]
Abstract
Repetitive transcranial magnetic stimulation (rTMS) emerged as a non-invasive brain stimulation technique in the treatment of psychiatric disorders. Both preclinical and clinical studies as well as systematic reviews provide a heterogeneous picture, particularly concerning the stimulation protocols used in rTMS. Here, we present a review of rTMS effects in rodent models of depressive-like symptoms with the aim to identify the most relevant factors that lead to an increased therapeutic success. The influence of different factors, such as the stimulation parameters (stimulus frequency and intensity, duration of stimulation, shape and positioning of the coil), symptom severity and individual characteristics (age, species and genetic background of the rodents), on the therapeutic success are discussed. Accumulating evidence indicates that rTMS ameliorates a multitude of depressive-like symptoms in rodent models, most effectively at high stimulation frequencies (≥5 Hz) especially in adult rodents with a pronounced pathological phenotype. The therapeutic success of rTMS might be increased in the future by considering these factors and using more standardized stimulation protocols.
Collapse
Affiliation(s)
- Iulia Zoicas
- Friedrich-Alexander University Erlangen-Nürnberg (FAU), Department of Psychiatry and Psychotherapy, Schwabachanlage 6, Erlangen 91054, Germany.
| | - Christiane Licht
- Paracelsus Medical University, Department of Psychiatry and Psychotherapy, Prof.-Ernst-Nathan-Str. 1, Nürnberg 90419, Germany
| | - Christiane Mühle
- Friedrich-Alexander University Erlangen-Nürnberg (FAU), Department of Psychiatry and Psychotherapy, Schwabachanlage 6, Erlangen 91054, Germany
| | - Johannes Kornhuber
- Friedrich-Alexander University Erlangen-Nürnberg (FAU), Department of Psychiatry and Psychotherapy, Schwabachanlage 6, Erlangen 91054, Germany
| |
Collapse
|
32
|
Darwish M, El Hajj R, Khayat L, Alaaeddine N. Stem Cell Secretions as a Potential Therapeutic Agent for Autism Spectrum Disorder: A Narrative Review. Stem Cell Rev Rep 2024; 20:1252-1272. [PMID: 38630359 DOI: 10.1007/s12015-024-10724-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2024] [Indexed: 07/04/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental illness characterized by impaired social interaction and restricted repetitive behaviors or interests. The rising prevalence of ASD diagnosis has triggered a surge in research into investigating the underlying neuropathological processes and finding new therapeutic approaches. ASD is characterized by neuroinflammation and dysregulation of neuro-immune cross-talk, which suggests that stem cell treatment might be a potential therapeutic approach. The beneficial and restorative effects of stem cells are mainly due to their paracrine activity, in which stem cells generate and release extracellular vesicles such as exosomes and distinct secreted non-vesicle soluble proteins, including, growth factors, chemokines, cytokines, and immunomodulatory molecules referred to as the Secretome. In this paper, we reviewed the existing research exploring the therapeutic potential of stem cell secretome focusing on their role in addressing ASD pathology. Furthermore, we proposed a comprehensive mechanism of action for stem cell secretions, encompassing the broader secretome as well as the specific contribution of exosomes, in alleviating ASD neuropathology. Across the reviewed studies, exosomes and secreted soluble factors of the transplanted stem cell demonstrate a potential efficacy in ameliorating autistic-like behaviors. The proposed mechanism of action involves the modulation of signaling pathways implicated in neuroinflammation, angiogenesis, cellular apoptosis, and immunomodulation.
Collapse
Affiliation(s)
- Mariam Darwish
- Faculty of Medical Sciences, Neuroscience Research Center, Lebanese University, Beirut, Lebanon
| | | | | | - Nada Alaaeddine
- Dean of Health Sciences, Modern University for Business & Science, Beirut, Lebanon.
| |
Collapse
|
33
|
Martín-Guerrero SM, Martín-Estebané M, Lara Ordóñez AJ, Cánovas M, Martín-Oliva D, González-Maeso J, Cutillas PR, López-Giménez JF. Maternal Immune Activation imprints translational dysregulation and differential MAP2 phosphorylation in descendant neural stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.07.597886. [PMID: 38895311 PMCID: PMC11185659 DOI: 10.1101/2024.06.07.597886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Alterations induced by maternal immune activation (MIA) during gestation impact the subsequent neurodevelopment of progeny, a process that in humans, has been linked to the development of several neuropsychiatric conditions. To undertake a comprehensive examination of the molecular mechanisms governing MIA, we have devised an in vitro model based on neural stem cells (NSCs) sourced from fetuses carried by animals subjected to Poly I:C treatment. These neural progenitors demonstrate proliferative capacity and can be effectively differentiated into both neurons and glial cells. Transcriptomic, proteomic, and phosphoproteomic analyses conducted on these cellular models, in conjunction with counterparts from control treatments, revealed discernible shifts in the expression levels of a specific subset of proteins implicated in neuronal function. Noteworthy, we found an absence of congruence between these alterations at the transcriptomic level, suggesting that differences in protein translation contribute to the observed dysregulation. Furthermore, the phosphoproteomic data highlighted a discernible discrepancy in the basal phosphorylation of proteins between differentiated cells from both experimental groups, particularly within proteins associated with cytoskeletal architecture and synaptic functionality, notably those belonging to the MAP family. Observed alterations in MAP phosphorylation were found to potentially have functional consequences as they correlate with changes in neuronal plasticity and the establishment of neuronal synapses. Our data agrees with previous published observations and further underscore the importance of MAP2 phosphorylation state on its function and the impact that this protein has in neuronal structure and function.
Collapse
|
34
|
Martz J, Shelton MA, Geist L, Seney ML, Kentner AC. Sex differences in offspring risk and resilience following 11β-hydroxylase antagonism in a rodent model of maternal immune activation. Neuropsychopharmacology 2024; 49:1078-1090. [PMID: 38007547 PMCID: PMC11109257 DOI: 10.1038/s41386-023-01771-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/21/2023] [Accepted: 11/08/2023] [Indexed: 11/27/2023]
Abstract
Maternal immune activation (MIA) puts offspring at greater risk for neurodevelopmental disorders associated with impaired social behavior. While it is known that immune signaling through maternal, placental, and fetal compartments contributes to these phenotypical changes, it is unknown to what extent the stress response to illness is involved and how it can be harnessed for potential interventions. To this end, on gestational day 15, pregnant rat dams were administered the bacterial mimetic lipopolysaccharide (LPS; to induce MIA) alongside metyrapone, a clinically available 11β-hydroxylase (11βHSD) inhibitor used to treat hypercortisolism in pregnant, lactating, and neonatal populations. Maternal, placental, and fetal brain levels of corticosterone and placental 11βHSD enzymes type 1 and 2 were measured 3-hrs post treatment. Offspring social behaviors were evaluated across critical phases of development. MIA was associated with increased maternal, placental, and fetal brain corticosterone concentrations that were diminished with metyrapone exposure. Metyrapone protected against reductions in placental 11βHSD2 in males only, suggesting that less corticosterone was inactivated in female placentas. Behaviorally, metyrapone-exposure attenuated MIA-induced social disruptions in juvenile, adolescent, and adult males, while females were unaffected or performed worse. Metyrapone-exposure reversed MIA-induced transcriptional changes in monoamine-, glutamate-, and GABA-related genes in adult male ventral hippocampus, but not in females. Taken together, these findings illustrate that MIA-induced HPA responses act alongside the immune system to produce behavioral deficits. As a clinically available drug, the sex-specific benefits and constraints of metyrapone should be investigated further as a potential means of reducing neurodevelopmental risks due to gestational MIA.
Collapse
Affiliation(s)
- Julia Martz
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, 02115, USA
| | - Micah A Shelton
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, USA
| | - Laurel Geist
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, 02115, USA
| | - Marianne L Seney
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, USA
| | - Amanda C Kentner
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, 02115, USA.
| |
Collapse
|
35
|
Zhu ZH, Yin XY, Cai Y, Jia NN, Wang PJ, Qi Q, Hou WL, Man LJ, Hui L. Association between the HHEX polymorphism and delayed memory in first-episode schizophrenic patients. Schizophr Res Cogn 2024; 36:100304. [PMID: 38444400 PMCID: PMC10912683 DOI: 10.1016/j.scog.2024.100304] [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: 11/27/2023] [Revised: 01/23/2024] [Accepted: 02/20/2024] [Indexed: 03/07/2024]
Abstract
The hematopoietically-expressed homeobox gene (HHEX) played a critical role in regulating the immune system that the abnormality of which was involved in the psychopathology and cognitive deficits of psychiatric disorders. The aim of this study was to investigate the effect of HHEX rs1111875 polymorphism on the susceptibility and cognitive deficits of first-episode schizophrenic patients (FSP). We assessed cognitive function in 239 first-episode patients meeting DSM-IV for schizophrenia, and 368 healthy controls using the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS). The HHEX rs1111875 polymorphism was genotyped. Our results showed that the allelic and genotypic frequencies of HHEX rs1111875 polymorphism didn't differ between FSP and healthy controls (both p > 0.05) after adjusting for sex and age. Cognitive test scores in FSP were significantly lower than those in healthy controls on all scales (all p < 0.001) except for the visuospatial/constructional score (p > 0.05) after adjusting for covariates. There was a significant genotype (p < 0.05) rather than genotype × diagnosis (p > 0.05) effect on the delayed memory score after adjusting for covariates. The HHEX rs1111875 polymorphism was significantly associated with the delayed memory score in FSP (p < 0.05), but not in healthy controls (p > 0.05) after adjusting for covariates. Our findings supported that the HHEX rs1111875 polymorphism did not contribute to the susceptibility to FSP. However, this polymorphism might influence the delayed memory in FSP. Moreover, FSP had poorer cognitive function than healthy controls except for the visuospatial/constructional domain.
Collapse
Affiliation(s)
| | | | | | - Ning Ning Jia
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Suzhou Medical College of Soochow University, Suzhou 215137, Jiangsu, PR China
| | - Pei Jie Wang
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Suzhou Medical College of Soochow University, Suzhou 215137, Jiangsu, PR China
| | - Qi Qi
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Suzhou Medical College of Soochow University, Suzhou 215137, Jiangsu, PR China
| | - Wen Long Hou
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Suzhou Medical College of Soochow University, Suzhou 215137, Jiangsu, PR China
| | - Li Juan Man
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Suzhou Medical College of Soochow University, Suzhou 215137, Jiangsu, PR China
| | - Li Hui
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Suzhou Medical College of Soochow University, Suzhou 215137, Jiangsu, PR China
| |
Collapse
|
36
|
Debs SR, Conn I, Navaneethan B, Penklis AG, Meyer U, Killcross S, Weickert CS, Purves-Tyson TD. Maternal immune activation and estrogen receptor modulation induce sex-specific dopamine-related behavioural and molecular alterations in adult rat offspring. Brain Behav Immun 2024; 118:236-251. [PMID: 38431238 DOI: 10.1016/j.bbi.2024.02.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/08/2024] [Accepted: 02/27/2024] [Indexed: 03/05/2024] Open
Abstract
Dopamine dysregulation contributes to psychosis and cognitive deficits in schizophrenia that can be modelled in rodents by inducing maternal immune activation (MIA). The selective estrogen receptor (ER) modulator, raloxifene, can improve psychosis and cognition in men and women with schizophrenia. However, few studies have examined how raloxifene may exert its therapeutic effects in mammalian brain in both sexes during young adulthood (age relevant to most prevalent age at diagnosis). Here, we tested the extent to which raloxifene alters dopamine-related behaviours and brain transcripts in young adult rats, both control and MIA-exposed females and males. We found that raloxifene increased amphetamine (AMPH)-induced locomotor activity in female controls, and in contrast, raloxifene reduced AMPH-induced locomotor activity in male MIA offspring. We did not detect overt prepulse inhibition (PPI) deficits in female or male MIA offspring, yet raloxifene enhanced PPI in male MIA offspring. Whereas, raloxifene ameliorated increased startle responsivity in female MIA offspring. In the substantia nigra (SN), we found reduced Drd2s mRNA in raloxifene-treated female offspring with or without MIA, and increased Comt mRNA in placebo-treated male MIA offspring relative to placebo-treated controls. These data demonstrate an underlying dopamine dysregulation in MIA animals that can become more apparent with raloxifene treatment, and may involve selective alterations in dopamine receptor levels and dopamine breakdown processes in the SN. Our findings support sex-specific, differential behavioural responses to ER modulation in MIA compared to control offspring, with beneficial effects of raloxifene treatment on dopamine-related behaviours relevant to schizophrenia found in male MIA offspring only.
Collapse
Affiliation(s)
- Sophie R Debs
- Preclinical Neuropsychiatry Laboratory, Neuroscience Research Australia, Sydney, Australia; Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, Australia; Discipline of Psychiatry & Mental Health, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Illya Conn
- Preclinical Neuropsychiatry Laboratory, Neuroscience Research Australia, Sydney, Australia; Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, Australia
| | - Brendan Navaneethan
- Preclinical Neuropsychiatry Laboratory, Neuroscience Research Australia, Sydney, Australia
| | - Andriane G Penklis
- Preclinical Neuropsychiatry Laboratory, Neuroscience Research Australia, Sydney, Australia; Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, Australia
| | - Urs Meyer
- Institute of Pharmacology and Toxicology, University of Zürich-Vetsuisse, Zürich, Switzerland; Switzerland Neuroscience Centre Zürich, Zürich, Switzerland
| | - Simon Killcross
- School of Psychology, University of New South Wales, Sydney, Australia
| | - Cynthia Shannon Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, Australia; Discipline of Psychiatry & Mental Health, Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Neuroscience & Physiology, Upstate Medical University, Syracuse, USA
| | - Tertia D Purves-Tyson
- Preclinical Neuropsychiatry Laboratory, Neuroscience Research Australia, Sydney, Australia; Discipline of Psychiatry & Mental Health, Faculty of Medicine, University of New South Wales, Sydney, Australia.
| |
Collapse
|
37
|
Chen RJ, Nabila A, Gal Toth J, Stuhlmann H, Toth M. The chemokine XCL1 functions as a pregnancy hormone to program offspring innate anxiety. Brain Behav Immun 2024; 118:178-189. [PMID: 38428650 PMCID: PMC11044916 DOI: 10.1016/j.bbi.2024.02.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 02/21/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024] Open
Abstract
Elevated levels of cytokines in maternal circulation increase the offspring's risk for neuropsychiatric disease. Because of their low homeostatic levels, circulating maternal cytokines during normal pregnancies have not been considered to play a role in fetal brain development and offspring behavior. Here we report that the T/NK cell chemotactic cytokine XCL1, a local paracrine immune signal, can function as a pregnancy hormone and is required for the proper development of placenta and male offspring approach-avoidance behavior. We found that circulating XCL1 levels were at a low pregestational level throughout pregnancy except for a midgestational rise and fall. Blunted elevation in maternal plasma XCL1 in dams with a genetic 5HT1A receptor deficit or following neutralization by anti-XCL1 antibodies increased the expression of tissue damage associated factors in WT fetal placenta and led to increased innate anxiety and stress reactivity in the WT male offspring. Therefore, chemokines like XCL1 may act as pregnancy hormones to regulate placenta development and offspring emotional behavior.
Collapse
Affiliation(s)
- Rosa J Chen
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Anika Nabila
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Judit Gal Toth
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Heidi Stuhlmann
- Cell and Developmental Biology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Miklos Toth
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
38
|
Zambon A, Rico LC, Herman M, Gundacker A, Telalovic A, Hartenberger LM, Kuehn R, Romanov RA, Hussaini SA, Harkany T, Pollak DD. Gestational immune activation disrupts hypothalamic neurocircuits of maternal care behavior. Mol Psychiatry 2024; 29:859-873. [PMID: 35581295 PMCID: PMC9112243 DOI: 10.1038/s41380-022-01602-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 12/27/2022]
Abstract
Immune activation is one of the most common complications during pregnancy, predominantly evoked by viral infections. Nevertheless, how immune activation affects mother-offspring relationships postpartum remains unknown. Here, by using the polyinosinic-polycytidylic acid (Poly I:C) model of gestational infection we show that viral-like immune activation at mid-gestation persistently changes hypothalamic neurocircuit parameters in mouse dams and, consequently, is adverse to parenting behavior. Poly I:C-exposed dams favor non-pup-directed exploratory behavior at the expense of pup retrieval. These behavioral deficits are underlain by dendrite pruning and lesser immediate early gene activation in Galanin (Gal)+ neurons with dam-specific transcriptional signatures that reside in the medial preoptic area (mPOA). Reduced activation of an exclusively inhibitory contingent of these distal-projecting Gal+ neurons allows for increased feed-forward inhibition onto putative dopaminergic neurons in the ventral tegmental area (VTA) in Poly I:C-exposed dams. Notably, destabilized VTA output specifically accompanies post-pup retrieval epochs. We suggest that gestational immunogenic insults bias both threat processing and reward perception, manifesting as disfavored infant caregiving.
Collapse
Affiliation(s)
- Alice Zambon
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Laura Cuenca Rico
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Mathieu Herman
- Department of Pathology and Cell Biology, Taub Institute, Columbia University Irving Medical Center, New York, NY, USA
| | - Anna Gundacker
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Amina Telalovic
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Lisa-Marie Hartenberger
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Rebekka Kuehn
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Roman A Romanov
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - S Abid Hussaini
- Department of Pathology and Cell Biology, Taub Institute, Columbia University Irving Medical Center, New York, NY, USA
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Daniela D Pollak
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
39
|
Hughes HK, Moreno RJ, Ashwood P. Innate Immune Dysfunction and Neuroinflammation in Autism Spectrum Disorder (ASD). FOCUS (AMERICAN PSYCHIATRIC PUBLISHING) 2024; 22:229-241. [PMID: 38680981 PMCID: PMC11046725 DOI: 10.1176/appi.focus.24022004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Autism spectrum disorder (ASD) is a highly heterogeneous neurodevelopmental disorder characterized by communication and social behavior deficits. The presence of restricted and repetitive behaviors often accompanies these deficits, and these characteristics can range from mild to severe. The past several decades have seen a significant rise in the prevalence of ASD. The etiology of ASD remains unknown; however, genetic and environmental risk factors play a role. Multiple hypotheses converge to suggest that neuroinflammation, or at least the interaction between immune and neural systems, may be involved in the etiology of some ASD cases or groups. Repeated evidence of innate immune dysfunction has been seen in ASD, often associated with worsening behaviors. This evidence includes data from circulating myeloid cells and brain resident macrophages/microglia in both human and animal models. This comprehensive review presents recent findings of innate immune dysfunction in ASD, including aberrant innate cellular function, evidence of neuroinflammation, and microglia activation. Appeared originally in Brain Behav Immun 2023; 108:245-254.
Collapse
Affiliation(s)
- H K Hughes
- Department of Medical Microbiology and Immunology, UC Davis, CA, USA (all authors);The M.I.N.D. Institute, University of California at Davis, CA, USA (all authors)
| | - R J Moreno
- Department of Medical Microbiology and Immunology, UC Davis, CA, USA (all authors);The M.I.N.D. Institute, University of California at Davis, CA, USA (all authors)
| | - P Ashwood
- Department of Medical Microbiology and Immunology, UC Davis, CA, USA (all authors);The M.I.N.D. Institute, University of California at Davis, CA, USA (all authors)
| |
Collapse
|
40
|
Li X, Fu Q, Zhong M, Long Y, Zhao F, Huang Y, Zhang Z, Wen M, Chen K, Chen R, Ma X. Quantitative proteomics of the miR-301a/SOCS3/STAT3 axis reveals underlying autism and anxiety-like behavior. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102136. [PMID: 38439911 PMCID: PMC10909786 DOI: 10.1016/j.omtn.2024.102136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/30/2024] [Indexed: 03/06/2024]
Abstract
Autism is a widespread neurodevelopmental disorder. Although the research on autism spectrum disorders has been increasing in the past decade, there is still no specific answer to its mechanism of action and treatment. As a pro-inflammatory microRNA, miR-301a is abnormally expressed in various psychiatric diseases including autism. Here, we show that miR-301a deletion and inhibition exhibited two distinct abnormal behavioral phenotypes in mice. We observed that miR-301a deletion in mice impaired learning/memory, and enhanced anxiety. On the contrary, miR-301a inhibition effectively reduced the maternal immune activation (MIA)-induced autism-like behaviors in mice. We further demonstrated that miR-301a bound to the 3'UTR region of the SOCS3, and that inhibition of miR-301a led to the upregulation of SOCS3 in hippocampus. The last result in the reduction of the inflammatory response by inhibiting phosphorylation of AKT and STAT3, and the expression level of IL-17A in poly(I:C)-induced autism-like features in mice. The obtained data revealed the miR-301a as a critical participant in partial behavior phenotypes, which may exhibit a divergent role between gene knockout and knockdown. Our findings ascertain that miR-301a negatively regulates SOCS3 in MIA-induced autism in mice and could present a new therapeutic target for ameliorating the behavioral abnormalities of autism.
Collapse
Affiliation(s)
- Xun Li
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China; Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou 510631, China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Qi Fu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China; Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou 510631, China
| | - Mingtian Zhong
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China; Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou 510631, China
| | - Yihao Long
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China; Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou 510631, China
| | - Fengyun Zhao
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China; Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou 510631, China
| | - Yanni Huang
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China; Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou 510631, China
| | - Zizhu Zhang
- Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Min Wen
- Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Kaizhao Chen
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China; Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou 510631, China
| | - Rongqing Chen
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Xiaodong Ma
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China; Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou 510631, China
| |
Collapse
|
41
|
Weickert TW, Ji E, Galletly C, Boerrigter D, Morishima Y, Bruggemann J, Balzan R, O’Donnell M, Liu D, Lenroot R, Weickert CS, Kindler J. Toll-Like Receptor mRNA Levels in Schizophrenia: Association With Complement Factors and Cingulate Gyrus Cortical Thinning. Schizophr Bull 2024; 50:403-417. [PMID: 38102721 PMCID: PMC10919782 DOI: 10.1093/schbul/sbad171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
BACKGROUND AND HYPOTHESES Previous studies revealed innate immune system activation in people with schizophrenia (SZ), potentially mediated by endogenous pathogen recognition receptors, notably Toll-like receptors (TLR). TLRs are activated by pathogenic molecules like bacterial lipopolysaccharides (TLR1 and TLR4), viral RNA (TLR3), or both (TLR8). Furthermore, the complement system, another key component of innate immunity, has previously been linked to SZ. STUDY DESIGN Peripheral mRNA levels of TLR1, TLR3, TLR4, and TLR8 were compared between SZ and healthy controls (HC). We investigated their relationship with immune activation through complement expression and cortical thickness of the cingulate gyrus, a region susceptible to immunological hits. TLR mRNA levels and peripheral complement receptor mRNA were extracted from 86 SZ and 77 HC white blood cells; structural MRI scans were conducted on a subset. STUDY RESULTS We found significantly higher TLR4 and TLR8 mRNA levels and lower TLR3 mRNA levels in SZ compared to HC. TLRs and complemental factors were significantly associated in SZ and HC, with the strongest deviations of TLR mRNA levels in the SZ subgroup having elevated complement expression. Cortical thickness of the cingulate gyrus was inversely associated with TLR8 mRNA levels in SZ, and with TLR4 and TLR8 levels in HC. CONCLUSIONS The study underscores the role of innate immune activation in schizophrenia, indicating a coordinated immune response of TLRs and the complement system. Our results suggest there could be more bacterial influence (based on TLR 4 levels) as opposed to viral influence (based on TLR3 levels) in schizophrenia. Specific TLRs were associated with brain cortical thickness reductions of limbic brain structures.
Collapse
Affiliation(s)
- Thomas W Weickert
- Neuroscience Research Australia, Schizophrenia Research Institute, Randwick, NSW 2031, Australia
- School of Psychiatry, University of New South Wales, Randwick, NSW 2031Australia
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY 13210, USA
| | - Ellen Ji
- Psychiatric University Hospital Zurich, Zurich, Switzerland
- Neuroscience Research Australia, Sydney, NSW, Australia
| | - Cherrie Galletly
- Discipline of Psychiatry, School of Medicine, University of Adelaide, Adelaide, SA, Australia
- Ramsay Health Care (SA) Mental Health, Adelaide, Australia
- Northern Adelaide Local Health Network, Adelaide, SA, Australia
| | - Danny Boerrigter
- Neuroscience Research Australia, Schizophrenia Research Institute, Randwick, NSW 2031, Australia
| | - Yosuke Morishima
- Translational Research Center, University Hospital of Psychiatry, University of Bern, Bern, Switzerland
| | - Jason Bruggemann
- Neuroscience Research Australia, Schizophrenia Research Institute, Randwick, NSW 2031, Australia
- School of Psychiatry, University of New South Wales, Randwick, NSW 2031Australia
- Edith Collins Centre (Translational Research in Alcohol Drugs and Toxicology), Sydney Local Health District, Sydney, Australia
- Speciality of Addiction Medicine, Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Ryan Balzan
- School of Psychology, Flinders University, Adelaide, SA, Australia
| | - Maryanne O’Donnell
- Neuroscience Research Australia, Schizophrenia Research Institute, Randwick, NSW 2031, Australia
- Kiloh Centre, Prince of Wales Hospital, Randwick, New South Wales, Australia
| | - Dennis Liu
- Discipline of Psychiatry, School of Medicine, University of Adelaide, Adelaide, SA, Australia
- Ramsay Health Care (SA) Mental Health, Adelaide, Australia
- Northern Adelaide Local Health Network, Adelaide, SA, Australia
| | - Rhoshel Lenroot
- Neuroscience Research Australia, Schizophrenia Research Institute, Randwick, NSW 2031, Australia
- School of Psychiatry, University of New South Wales, Randwick, NSW 2031Australia
- Department of Psychiatry, University of New Mexico, Albuquerque, NM 87131-0001, USA
| | - Cynthia Shannon Weickert
- Neuroscience Research Australia, Schizophrenia Research Institute, Randwick, NSW 2031, Australia
- School of Psychiatry, University of New South Wales, Randwick, NSW 2031Australia
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY 13210, USA
| | - Jochen Kindler
- Neuroscience Research Australia, Schizophrenia Research Institute, Randwick, NSW 2031, Australia
- University Hospital of Child and Adolescent Psychiatry and Psychotherapy, University of Bern, 3000 Bern, Switzerland
| |
Collapse
|
42
|
Nielsen TC, Nassar N, Shand AW, Jones HF, Han VX, Patel S, Guastella AJ, Dale RC, Lain SJ. Association between cumulative maternal exposures related to inflammation and child attention-deficit/hyperactivity disorder: A cohort study. Paediatr Perinat Epidemiol 2024; 38:241-250. [PMID: 38009577 DOI: 10.1111/ppe.13022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/10/2023] [Accepted: 11/11/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Preclinical studies suggest synergistic effects of maternal inflammatory exposures on offspring neurodevelopment, but human studies have been limited. OBJECTIVES To examine the cumulative association and potential interactions between seven maternal exposures related to inflammation and child attention-deficit/hyperactivity disorder (ADHD). METHODS We conducted a population-based cohort study of children born from July 2001 to December 2011 in New South Wales, Australia, and followed up until December 2014. Seven maternal exposures were identified from birth data and hospital admissions during pregnancy: autoimmune disease, asthma, hospitalization for infection, mood or anxiety disorder, smoking, hypertension, and diabetes. Child ADHD was identified from stimulant prescription records. Multivariable Cox regression assessed the association between individual and cumulative exposures and ADHD and potential interaction between exposures, controlling for potential confounders. RESULTS The cohort included 908,770 children, one-third (281,724) with one or more maternal exposures. ADHD was identified in 16,297 children (incidence 3.5 per 1000 person-years) with median age of 7 (interquartile range 2) years at first treatment. Each exposure was independently associated with ADHD, and risk increased with additional exposures: one exposure (hazard ratio (HR) 1.59, 95% confidence interval (CI) 1.54, 1.65), two exposures (HR 2.25, 95% CI 2.13, 2.37), and three or more exposures (HR 3.28, 95% CI 2.95, 3.64). Positive interaction was found between smoking and infection. The largest effect size was found for cumulative exposure of asthma, infection, mood or anxiety disorder, and smoking (HR 6.12, 95% CI 3.47, 10.70). CONCLUSIONS This study identifies cumulative effects of multiple maternal exposures related to inflammation on ADHD, most potentially preventable or modifiable. Future studies should incorporate biomarkers of maternal inflammation and consider gene-environment interactions.
Collapse
Affiliation(s)
- Timothy C Nielsen
- Children's Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Natasha Nassar
- Children's Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Antonia W Shand
- Children's Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Royal Hospital for Women, Randwick, New South Wales, Australia
| | - Hannah F Jones
- Children's Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Starship Children's Hospital, Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Velda X Han
- Children's Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore City, Singapore
| | - Shrujna Patel
- Children's Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Adam J Guastella
- Children's Hospital Westmead Clinical School, Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Russell C Dale
- Children's Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Samantha J Lain
- Children's Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
43
|
Jaswa EG, Cedars MI, Lindquist KJ, Bishop SL, Kim YS, Kaing A, Prahl M, Gaw SL, Corley J, Hoskin E, Cho YJ, Rogers E, Huddleston HG. In Utero Exposure to Maternal COVID-19 Vaccination and Offspring Neurodevelopment at 12 and 18 Months. JAMA Pediatr 2024; 178:258-265. [PMID: 38252445 PMCID: PMC10804280 DOI: 10.1001/jamapediatrics.2023.5743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 10/26/2023] [Indexed: 01/23/2024]
Abstract
Importance Uptake of COVID-19 vaccines among pregnant individuals was hampered by safety concerns around potential risks to unborn children. Data clarifying early neurodevelopmental outcomes of offspring exposed to COVID-19 vaccination in utero are lacking. Objective To determine whether in utero exposure to maternal COVID-19 vaccination was associated with differences in scores on the Ages and Stages Questionnaire, third edition (ASQ-3), at 12 and 18 months of age. Design, Setting, and Participants This prospective cohort study, Assessing the Safety of Pregnancy During the Coronavirus Pandemic (ASPIRE), enrolled pregnant participants from May 2020 to August 2021; follow-up of children from these pregnancies is ongoing. Participants, which included pregnant individuals and their offspring from all 50 states, self-enrolled online. Study activities were performed remotely. Exposure In utero exposure of the fetus to maternal COVID-19 vaccination during pregnancy was compared with those unexposed. Main Outcomes and Measures Neurodevelopmental scores on validated ASQ-3, completed by birth mothers at 12 and 18 months. A score below the established cutoff in any of 5 subdomains (communication, gross motor, fine motor, problem solving, social skills) constituted an abnormal screen for developmental delay. Results A total of 2487 pregnant individuals (mean [SD] age, 33.3 [4.2] years) enrolled at less than 10 weeks' gestation and completed research activities, yielding a total of 2261 and 1940 infants aged 12 and 18 months, respectively, with neurodevelopmental assessments. In crude analyses, 471 of 1541 exposed infants (30.6%) screened abnormally for developmental delay at 12 months vs 203 of 720 unexposed infants (28.2%; χ2 = 1.32; P = .25); the corresponding prevalences at 18 months were 262 of 1301 (20.1%) vs 148 of 639 (23.2%), respectively (χ2 = 2.35; P = .13). In multivariable mixed-effects logistic regression models adjusting for maternal age, race, ethnicity, education, income, maternal depression, and anxiety, no difference in risk for abnormal ASQ-3 screens was observed at either time point (12 months: adjusted risk ratio [aRR], 1.14; 95% CI, 0.97-1.33; 18 months: aRR, 0.88; 95% CI, 0.72-1.07). Further adjustment for preterm birth and infant sex did not affect results (12 months: aRR, 1.16; 95% CI, 0.98-1.36; 18 months: aRR, 0.87; 95% CI, 0.71-1.07). Conclusions and Relevance Results of this cohort study suggest that COVID-19 vaccination was safe during pregnancy from the perspective of infant neurodevelopment to 18 months of age. Additional longer-term research should be conducted to corroborate these findings and buttress clinical guidance with a strong evidence base.
Collapse
Affiliation(s)
- Eleni G. Jaswa
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Endocrinology and Infertility, University of California, San Francisco, San Francisco
| | - Marcelle I. Cedars
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Endocrinology and Infertility, University of California, San Francisco, San Francisco
| | - Karla J. Lindquist
- Department of Epidemiology & Biostatistics, University of California, San Francisco, San Francisco
| | - Somer L. Bishop
- Department of Psychiatry, University of California, San Francisco, San Francisco
| | - Young-Shin Kim
- Department of Psychiatry, University of California, San Francisco, San Francisco
| | - Amy Kaing
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Endocrinology and Infertility, University of California, San Francisco, San Francisco
| | - Mary Prahl
- Department of Pediatrics, Division of Pediatric Infectious Disease and Global Health, University of California, San Francisco, San Francisco
| | - Stephanie L. Gaw
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Maternal Fetal Medicine, University of California, San Francisco, San Francisco
| | - Jamie Corley
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Endocrinology and Infertility, University of California, San Francisco, San Francisco
| | - Elena Hoskin
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Endocrinology and Infertility, University of California, San Francisco, San Francisco
| | - Yoon Jae Cho
- Department of Psychiatry, University of California, San Francisco, San Francisco
| | - Elizabeth Rogers
- Department of Pediatrics, Division of Neonatology, University of California, San Francisco, San Francisco
| | - Heather G. Huddleston
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Endocrinology and Infertility, University of California, San Francisco, San Francisco
| |
Collapse
|
44
|
Zhang HL, Hu S, Qu ST, Lv MD, Wang JJ, Liu XT, Yao JH, Ding YY, Xu GY. Inhibition of NKCC1 Ameliorates Anxiety and Autistic Behaviors Induced by Maternal Immune Activation in Mice. Curr Issues Mol Biol 2024; 46:1851-1864. [PMID: 38534737 DOI: 10.3390/cimb46030121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/28/2024] Open
Abstract
Autism spectrum disorder (ASD) is thought to result from susceptibility genotypes and environmental risk factors. The offspring of women who experience pregnancy infection have an increased risk for autism. Maternal immune activation (MIA) in pregnant animals produces offspring with autistic behaviors, making MIA a useful model for autism. However, how MIA causes autistic behaviors in offspring is not fully understood. Here, we show that NKCC1 is critical for mediating autistic behaviors in MIA offspring. We confirmed that MIA induced by poly(I:C) infection during pregnancy leads to autistic behaviors in offspring. We further demonstrated that MIA offspring showed significant microglia activation, excessive dendritic spines, and narrow postsynaptic density (PSD) in their prefrontal cortex (PFC). Then, we discovered that these abnormalities may be caused by overexpression of NKCC1 in MIA offspring's PFCs. Finally, we ameliorated the autistic behaviors using PFC microinjection of NKCC1 inhibitor bumetanide (BTN) in MIA offspring. Our findings may shed new light on the pathological mechanisms for autism caused by pregnancy infection.
Collapse
Affiliation(s)
- Hai-Long Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Suzhou Medical College of Soochow University, Medical Center of Soochow University, Suzhou 215123, China
| | - Shufen Hu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Suzhou Medical College of Soochow University, Medical Center of Soochow University, Suzhou 215123, China
| | - Shu-Ting Qu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Suzhou Medical College of Soochow University, Medical Center of Soochow University, Suzhou 215123, China
| | - Meng-Dan Lv
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Suzhou Medical College of Soochow University, Medical Center of Soochow University, Suzhou 215123, China
| | - Jun-Jun Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Suzhou Medical College of Soochow University, Medical Center of Soochow University, Suzhou 215123, China
| | - Xin-Ting Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Suzhou Medical College of Soochow University, Medical Center of Soochow University, Suzhou 215123, China
| | - Jia-He Yao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Suzhou Medical College of Soochow University, Medical Center of Soochow University, Suzhou 215123, China
| | - Yi-Yan Ding
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Suzhou Medical College of Soochow University, Medical Center of Soochow University, Suzhou 215123, China
| | - Guang-Yin Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Suzhou Medical College of Soochow University, Medical Center of Soochow University, Suzhou 215123, China
| |
Collapse
|
45
|
DelaCuesta-Barrutia J, Martínez-Peula O, Rivero G, Santas-Martín JA, Munarriz-Cuezva E, Brocos-Mosquera I, Miranda-Azpiazu P, Diez-Alarcia R, Morentin B, Honer WG, Callado LF, Erdozain AM, Ramos-Miguel A. Effect of antipsychotic drugs on group II metabotropic glutamate receptor expression and epigenetic control in postmortem brains of schizophrenia subjects. Transl Psychiatry 2024; 14:113. [PMID: 38396013 PMCID: PMC10891050 DOI: 10.1038/s41398-024-02832-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Antipsychotic-induced low availability of group II metabotropic glutamate receptors (including mGlu2R and mGlu3R) in brains of schizophrenia patients may explain the limited efficacy of mGlu2/3R ligands in clinical trials. Studies evaluating mGlu2/3R levels in well-designed, large postmortem brain cohorts are needed to address this issue. Postmortem samples from the dorsolateral prefrontal cortex of 96 schizophrenia subjects and matched controls were collected. Toxicological analyses identified cases who were (AP+) or were not (AP-) receiving antipsychotic treatment near the time of death. Protein and mRNA levels of mGlu2R and mGlu3R, as well as GRM2 and GRM3 promoter-attached histone posttranslational modifications, were quantified. Experimental animal models were used to compare with data obtained in human tissues. Compared to matched controls, schizophrenia cortical samples had lower mGlu2R protein amounts, regardless of antipsychotic medication. Downregulation of mGlu3R was observed in AP- schizophrenia subjects only. Greater predicted occupancy values of dopamine D2 and serotonin 5HT2A receptors correlated with higher density of mGlu3R, but not mGlu2R. Clozapine treatment and maternal immune activation in rodents mimicked the mGlu2R, but not mGlu3R regulation observed in schizophrenia brains. mGlu2R and mGlu3R mRNA levels, and the epigenetic control mechanisms did not parallel the alterations at the protein level, and in some groups correlated inversely. Insufficient cortical availability of mGlu2R and mGlu3R may be associated with schizophrenia. Antipsychotic treatment may normalize mGlu3R, but not mGlu2R protein levels. A model in which epigenetic feedback mechanisms controlling mGlu3R expression are activated to counterbalance mGluR loss of function is described.
Collapse
Affiliation(s)
| | | | - Guadalupe Rivero
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Leioa, Spain
| | - Jon A Santas-Martín
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - Eva Munarriz-Cuezva
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Leioa, Spain
| | - Iria Brocos-Mosquera
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
| | | | - Rebeca Diez-Alarcia
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Leioa, Spain
| | - Benito Morentin
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Basque Institute of Legal Medicine, Bilbao, Spain
| | - William G Honer
- Department Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Luis F Callado
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Leioa, Spain
| | - Amaia M Erdozain
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Leioa, Spain
| | - Alfredo Ramos-Miguel
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain.
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Leioa, Spain.
| |
Collapse
|
46
|
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
|
47
|
Moreno-Fernández M, Ucha M, Reis-de-Paiva R, Marcos A, Ambrosio E, Higuera-Matas A. Lack of interactions between prenatal immune activation and Δ 9-tetrahydrocannabinol exposure during adolescence in behaviours relevant to symptom dimensions of schizophrenia in rats. Prog Neuropsychopharmacol Biol Psychiatry 2024; 129:110889. [PMID: 37918558 DOI: 10.1016/j.pnpbp.2023.110889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 07/27/2023] [Accepted: 10/29/2023] [Indexed: 11/04/2023]
Abstract
The causality in the association between cannabis use and the risk of developing schizophrenia has been the subject of intense debate in the last few years. The development of animal models recapitulating several aspects of the disease is crucial for shedding light on this issue. Given that maternal infections are a known risk for schizophrenia, here, we used the maternal immune activation (MIA) model combined with THC exposure during adolescence to examine several behaviours in rats (working memory in the Y maze, sociability in the three-chamber test, sucrose preference as a measure, prepulse inhibition and formation of incidental associations) that are similar to the different symptom clusters of the disease. To this end, we administered LPS to pregnant dams and when the offspring reached adolescence, we exposed them to a mild dose of THC to examine their behaviour in adulthood. We also studied several parameters in the dams, including locomotor activity in the open field, elevated plus maze performance and their response to LPS, that could predict symptom severity of the offspring, but found no evidence of any predictive value of these variables. In the adult offspring, MIA was associated with impaired working memory and sensorimotor gating, but surprisingly, it increased sociability, social novelty and sucrose preference. THC, on its own, impaired sociability and social memory, but there were no interactions between MIA and THC exposure. These results suggest that, in this model, THC during adolescence does not trigger or aggravate symptoms related to schizophrenia in rats.
Collapse
Affiliation(s)
- Mario Moreno-Fernández
- Department of Psychobiology, School of Psychology, National University for Distance Learning (UNED), Madrid, Spain; UNED International Graduate School (EIDUNED), Madrid, Spain
| | - Marcos Ucha
- Department of Psychobiology, School of Psychology, National University for Distance Learning (UNED), Madrid, Spain.
| | - Raquel Reis-de-Paiva
- Department of Psychobiology, School of Psychology, National University for Distance Learning (UNED), Madrid, Spain
| | - Alberto Marcos
- Department of Psychobiology, School of Psychology, National University for Distance Learning (UNED), Madrid, Spain
| | - Emilio Ambrosio
- Department of Psychobiology, School of Psychology, National University for Distance Learning (UNED), Madrid, Spain
| | - Alejandro Higuera-Matas
- Department of Psychobiology, School of Psychology, National University for Distance Learning (UNED), Madrid, Spain.
| |
Collapse
|
48
|
Norman TR. Cannabidiol, cognition and schizophrenia: a narrative review. EXPLORATION OF MEDICINE 2024; 5:48-58. [DOI: 10.37349/emed.2024.00204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/02/2023] [Indexed: 03/05/2025] Open
Abstract
Schizophrenia is a serious mental disorder affecting about 1% of the population. It is characterised by multiple symptoms which are mostly responsive to treatment with antipsychotic medications. Cognitive impairment is regarded as a core feature of illness which is mostly poorly responsive to treatment with the current antipsychotic medications. Improving cognitive function is an important treatment goal as it is associated with better outcomes in employment and quality of life. Adjunctive pharmacological treatments have been examined to improve measures of cognition but with limited success. Cannabidiol (CBD), has shown promise in preclinical models of cognitive deficits of schizophrenia. On the other hand, limited studies in small groups of patients with schizophrenia have shown no significant clinical benefits for cognitive function as an adjunct to ongoing treatment with antipsychotics. A single trial, in which CBD as a standalone treatment was compared to the antipsychotic medication amisulpride, showed significant changes in cognitive measures for both agents, with no statistically significant difference between them. It might therefore be concluded that the preclinical findings have failed to translate to the clinic. However, the preclinical findings themselves are based on a circumscribed set of studies in multiple cognitive models and have used varying doses and routes of drug administration. The same general methodological issues are present in the suite of clinical studies. Issues such as patient heterogeneity in terms of illness duration, formulation and dose of CBD employed, and length of cannabinoid treatment might militate positive findings. The limited clinical database available makes the benefits (or lack thereof) of CBD for the cognitive effects of schizophrenia uncertain. Continued research in much larger patient populations than have so far been investigated as well as a consideration of dose ranging studies are required to fully assess the potential risks against the benefits of CBD treatment for cognitive deficits in schizophrenia.
Collapse
Affiliation(s)
- Trevor R. Norman
- Department of Psychiatry, University of Melbourne, Heidelberg 3084, Australia
| |
Collapse
|
49
|
Stankovic I, Notaras M, Wolujewicz P, Lu T, Lis R, Ross ME, Colak D. Schizophrenia endothelial cells exhibit higher permeability and altered angiogenesis patterns in patient-derived organoids. Transl Psychiatry 2024; 14:53. [PMID: 38263175 PMCID: PMC10806043 DOI: 10.1038/s41398-024-02740-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 12/29/2023] [Accepted: 01/05/2024] [Indexed: 01/25/2024] Open
Abstract
Schizophrenia (SCZ) is a complex neurodevelopmental disorder characterized by the manifestation of psychiatric symptoms in early adulthood. While many research avenues into the origins of SCZ during brain development have been explored, the contribution of endothelial/vascular dysfunction to the disease remains largely elusive. To model the neuropathology of SCZ during early critical periods of brain development, we utilized patient-derived induced pluripotent stem cells (iPSCs) to generate 3D cerebral organoids and define cell-specific signatures of disease. Single-cell RNA sequencing revealed that while SCZ organoids were similar in their macromolecular diversity to organoids generated from healthy controls (CTRL), SCZ organoids exhibited a higher percentage of endothelial cells when normalized to total cell numbers. Additionally, when compared to CTRL, differential gene expression analysis revealed a significant enrichment in genes that function in vessel formation, vascular regulation, and inflammatory response in SCZ endothelial cells. In line with these findings, data from 23 donors demonstrated that PECAM1+ microvascular vessel-like structures were increased in length and number in SCZ organoids in comparison to CTRL organoids. Furthermore, we report that patient-derived endothelial cells displayed higher paracellular permeability, implicating elevated vascular activity. Collectively, our data identified altered gene expression patterns, vessel-like structural changes, and enhanced permeability of endothelial cells in patient-derived models of SCZ. Hence, brain microvascular cells could play a role in the etiology of SCZ by modulating the permeability of the developing blood brain barrier (BBB).
Collapse
Affiliation(s)
- Isidora Stankovic
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Michael Notaras
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Paul Wolujewicz
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Tyler Lu
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Raphael Lis
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY, USA
| | - M Elizabeth Ross
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Dilek Colak
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
50
|
Gonçalves CL, Doifode T, Rezende VL, Costa MA, Rhoads JM, Soutullo CA. The many faces of microbiota-gut-brain axis in autism spectrum disorder. Life Sci 2024; 337:122357. [PMID: 38123016 DOI: 10.1016/j.lfs.2023.122357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/02/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
The gut-brain axis is gaining more attention in neurodevelopmental disorders, especially autism spectrum disorder (ASD). Many factors can influence microbiota in early life, including host genetics and perinatal events (infections, mode of birth/delivery, medications, nutritional supply, and environmental stressors). The gut microbiome can influence blood-brain barrier (BBB) permeability, drug bioavailability, and social behaviors. Developing microbiota-based interventions such as probiotics, gastrointestinal (GI) microbiota transplantation, or metabolite supplementation may offer an exciting approach to treating ASD. This review highlights that RNA sequencing, metabolomics, and transcriptomics data are needed to understand how microbial modulators can influence ASD pathophysiology. Due to the substantial clinical heterogeneity of ASD, medical caretakers may be unlikely to develop a broad and effective general gut microbiota modulator. However, dietary modulation followed by administration of microbiota modulators is a promising option for treating ASD-related behavioral and gastrointestinal symptoms. Future work should focus on the accuracy of biomarker tests and developing specific psychobiotic agents tailored towards the gut microbiota seen in ASD patients, which may include developing individualized treatment options.
Collapse
Affiliation(s)
- Cinara L Gonçalves
- Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| | - Tejaswini Doifode
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health (UTHealth), Houston, TX, USA
| | - Victoria L Rezende
- Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Maiara A Costa
- Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - J Marc Rhoads
- Department of Pediatrics, Division of Pediatric Gastroenterology, McGovern Medical School, The University of Texas Health (UTHealth), Houston, TX, USA
| | - Cesar A Soutullo
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health (UTHealth), Houston, TX, USA
| |
Collapse
|